Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Immunol ; 202(5): 1521-1530, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30700584

ABSTRACT

Hypoxia is a common and prominent feature of the microenvironment at sites of bacteria-associated inflammation in inflammatory bowel disease. The prolyl-hydroxylases (PHD1/2/3) and the asparaginyl-hydroxylase factor-inhibiting HIF are oxygen-sensing enzymes that regulate adaptive responses to hypoxia through controlling the activity of HIF and NF-κB-dependent transcriptional pathways. Previous studies have demonstrated that the pan-hydroxylase inhibitor dimethyloxalylglycine (DMOG) is effective in the alleviation of inflammation in preclinical models of inflammatory bowel disease, at least in part, through suppression of IL-1ß-induced NF-κB activity. TLR-dependent signaling in immune cells, such as monocytes, which is important in bacteria-driven inflammation, shares a signaling pathway with IL-1ß. In studies into the effect of pharmacologic hydroxylase inhibition on TLR-induced inflammation in monocytes, we found that DMOG selectively triggers cell death in cultured THP-1 cells and primary human monocytes at concentrations well tolerated in other cell types. DMOG-induced apoptosis was independent of increased caspase-3/7 activity but was accompanied by reduced expression of the inhibitor of apoptosis protein 1 (cIAP1). Based on these data, we hypothesize that pharmacologic inhibition of the HIF-hydroxylases selectively targets monocytes for cell death and that this may contribute to the anti-inflammatory activity of HIF-hydroxylase inhibitors.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Inflammation/drug therapy , Mixed Function Oxygenases/antagonists & inhibitors , Monocytes/drug effects , Prolyl-Hydroxylase Inhibitors/pharmacology , Cell Death/drug effects , Cell Death/immunology , Cells, Cultured , HEK293 Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Mixed Function Oxygenases/immunology , Mixed Function Oxygenases/metabolism , Monocytes/immunology , Monocytes/metabolism
2.
Xenotransplantation ; 26(4): e12516, 2019 07.
Article in English | MEDLINE | ID: mdl-30989742

ABSTRACT

Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications. We suggest that a pig with nine genetic modifications (ie, currently available) will provide organs (initially kidneys and hearts) that would function for a clinically valuable period of time, for example, >12 months, after transplantation into patients with end-stage organ failure. The national regulatory authorities, however, will likely require evidence, based on in vitro and/or in vivo experimental data, to justify the inclusion of each individual genetic modification in the pig. We provide data both from our own experience and that of others on the advantages of pigs in which (a) all three known carbohydrate xenoantigens have been deleted (triple-knockout pigs), (b) two human complement-regulatory proteins (CD46, CD55) and two human coagulation-regulatory proteins (thrombomodulin, endothelial cell protein C receptor) are expressed, (c) the anti-apoptotic and "anti-inflammatory" molecule, human hemeoxygenase-1 is expressed, and (d) human CD47 is expressed to suppress elements of the macrophage and T-cell responses. Although many alternative genetic modifications could be made to an organ-source pig, we suggest that the genetic manipulations we identify above will all contribute to the success of the initial clinical pig kidney or heart transplants, and that the beneficial contribution of each individual manipulation is supported by considerable experimental evidence.


Subject(s)
Animals, Genetically Modified/genetics , Graft Rejection/prevention & control , Swine/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified/immunology , CD47 Antigen/genetics , CD47 Antigen/immunology , CD55 Antigens/genetics , CD55 Antigens/immunology , Endothelial Protein C Receptor/genetics , Endothelial Protein C Receptor/immunology , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knock-In Techniques , Gene Knockout Techniques , Graft Rejection/immunology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Humans , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , Swine/immunology , Thrombomodulin/genetics , Thrombomodulin/immunology
3.
Xenotransplantation ; 26(4): e12504, 2019 07.
Article in English | MEDLINE | ID: mdl-30825348

ABSTRACT

The humoral barrier has been the limiting factor in moving xenotransplantation towards the clinic. Improvements in somatic cell nuclear transfer and genome editing, particularly CRISPR-Cas9, have made it possible to create pigs with multiple glycan xenoantigen deletions for the purposes of reducing xenoreactive antibody binding to the xenografted organ. Recent studies have also considered the aetiology and existence of antibodies directed at the swine leucocyte antigen (SLA) complex, and potential genetic engineering strategies to avoid these antibodies. Evaluation of xenoreactive antibody binding is very important for the advancement of xenotransplantation, because if patients do not have any detectable xenoreactive antibody, then it is reasonable to expect that cellular rejection and not antibody-mediated rejection (AMR) will be the next hurdle to clinical application.


Subject(s)
Antigens, Heterophile/immunology , Galactosyltransferases/immunology , Gene Knockout Techniques , Graft Rejection/prevention & control , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/immunology , Swine/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified/immunology , Antibodies, Heterophile/biosynthesis , Antibodies, Heterophile/immunology , Antigen-Antibody Reactions , Antigens, Heterophile/genetics , Epitopes/immunology , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Genetic Engineering , Graft Rejection/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/genetics , Transplantation Immunology
4.
Acta Microbiol Immunol Hung ; 66(2): 219-234, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30484328

ABSTRACT

Immunogenic potency of the recombinant Erp, HspR, LppX, MmaA4, and OmpA proteins from Mycobacterium tuberculosis (MTB), formulated with Montanide ISA 720 VG adjuvant, was evaluated in BALB/c mice for the first time in this study. The five vaccine formulations, adjuvant, and BCG vaccine were subcutaneously injected into mice, and the sera were collected at days 0, 15, 30, 41, and 66. The humoral and cellular immune responses against vaccine formulations were determined by measuring serum IgG and serum interferon-gamma (IFN-γ) and interleukin-12 (IL-12) levels, respectively. All formulations significantly increased IgG levels post-vaccination. The highest increase in IFN-γ level was provided by MmaA4 formulation. The Erp, HspR, and LppX formulations were as effective as BCG in enhancement of IFN-γ level. The most efficient vaccine boosting the IL-12 level was HspR formulation, especially at day 66. Erp formulation also increased the IL-12 level more than BCG at days 15 and 30. The IL-12 level boosted by MmaA4 formulation was found to be similar to that by BCG. OmpA formulation was inefficient in enhancement of cellular immune responses. This study showed that MmaA4, HspR, and Erp proteins from MTB are successful in eliciting both humoral and cellular immune responses in mice.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Heat-Shock Proteins/immunology , Membrane Proteins/immunology , Methyltransferases/immunology , Mixed Function Oxygenases/immunology , Repressor Proteins/immunology , Tuberculosis Vaccines/immunology , Animals , Antibodies, Bacterial/blood , BCG Vaccine/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Cytokines/immunology , Female , Heat-Shock Proteins/genetics , Immunity, Cellular , Immunity, Humoral , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Membrane Proteins/genetics , Methyltransferases/genetics , Mice , Mice, Inbred BALB C , Mixed Function Oxygenases/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Oleic Acids/administration & dosage , Repressor Proteins/genetics , Tuberculosis/microbiology , Tuberculosis/prevention & control , Vaccination , Vaccines, Synthetic/immunology
5.
Int J Mol Sci ; 20(8)2019 Apr 24.
Article in English | MEDLINE | ID: mdl-31022963

ABSTRACT

Tet-eleven translocation 1 (TET1) is a dioxygenase that plays an important role in decreasing the abundance of DNA methylation and changing the expression levels of specific genes related to inflammation. Porphyromonas gingivalis (Pg.) lipopolysaccharide (LPS) can induce periodontal diseases that present with severe bone loss and collagen fiber destruction accompanied by a high number of M1 macrophages. M1-polarized macrophages are pivotal immune cells that promote the progression of the periodontal inflammatory response, but the function of TET1 during M1 macrophage activation is still unknown. Our results showed that the mRNA and protein expression levels of TET1 decreased in THP-1 cells during M1 macrophage differentiation. TET1 knockdown resulted in a significant decrease in the production of proinflammatory markers such as IL-6, TNF-α, CCL2, and HLA-DR in Pg. LPS/IFN-γ- and Escherichia coli (E. coli) LPS/IFN-γ-induced M1 macrophages. Mechanistically, TET1 knockdown downregulated the activity of the NF-κB signaling pathway. After treatment with the NF-κB inhibitor BAY 11-7082, M1 marker expression showed no significant difference between the TET1 knockdown group and the control group. Taken together, these results suggest that TET1 depletion inhibited Pg. LPS/IFN-γ-induced M1 macrophage polarization through the NF-κB pathway in THP-1 cells.


Subject(s)
Interferon-gamma/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Mixed Function Oxygenases/genetics , NF-kappa B/immunology , Porphyromonas gingivalis/immunology , Proto-Oncogene Proteins/genetics , Gene Knockdown Techniques , Humans , Inflammation/immunology , Inflammation/microbiology , Macrophage Activation , Macrophages/microbiology , Mixed Function Oxygenases/immunology , Proto-Oncogene Proteins/immunology , Signal Transduction , THP-1 Cells
6.
Int J Mol Sci ; 20(7)2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30925699

ABSTRACT

High population density alters insect prophylactic immunity, with density-dependent prophylaxis (DDP) being reported in many polyphonic insects. However, the molecular mechanism for DDP remains unclear. In current study, the role of tyramine ß-hydroxylase (Tßh) in the immune response of M. separata larvae that were subject to different rearing densities conditions was investigated. The tyramine ß-hydroxylase activity of larvae from high density treatments (10 and 30 larvae per jar) was significantly higher than that of the larvae from low density treatments (one, two, and five larvae/jar). A tyramine ß-hydroxylase (designated MsTßh) containing a 1779 bp open reading frame was identified. Multiple sequence alignment and phylogenetic analysis indicated that MsTßh was orthologous to the Tßh that was found in other lepidopterans. Elevated MsTßh expression was observed in larvae under high density (10 larvae per jar). Silencing MsTßh expression by the injection of dsRNA in larvae from the high density treatment produced a 25.1% reduction in octopamine levels, while at the same time, there was a significant decrease in phenoloxidase (PO) and lysozyme activity, total haemocyte counts, and survival against Beauveria infection 56.6%, 88.5%, 82.0%, and 55.8%, respectively, when compared to control larvae. Our findings provide the first insights into how MsTßh mediates the octopamine level, which in turn modulates the immune response of larvae under different population densities.


Subject(s)
Insect Proteins/immunology , Mixed Function Oxygenases/immunology , Moths/immunology , Amino Acid Sequence , Animals , Beauveria/immunology , Immunity , Insect Proteins/chemistry , Insect Proteins/genetics , Larva/chemistry , Larva/genetics , Larva/immunology , Larva/microbiology , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Moths/chemistry , Moths/genetics , Moths/microbiology , Phylogeny , Sequence Alignment
7.
Clin Immunol ; 196: 85-96, 2018 11.
Article in English | MEDLINE | ID: mdl-29894742

ABSTRACT

Here, we determined the 5-hydroxymethylcytosine (5hmC), 5-methylcytosine (5mC), Ten Eleven Translocation (TETs), and DNA methyltransferases (DNMTs) levels in epithelial and inflammatory cells of labial salivary glands (LSG) from Sjögren's syndrome (SS)-patients and the effect of cytokines on HSG cells. LSG from SS-patients, controls and HSG cells incubated with cytokines were analysed. Levels of 5mC, 5hmC, DNMTs, TET2 and MeCP2 were assessed by immunofluorescence. In epithelial cells from SS-patients, an increase in TET2, 5hmC and a decrease in 5mC and MeCP2 were observed, additionally, high levels of 5mC and DNMTs and low levels of 5hmC were detected in inflammatory cells. Cytokines increased TET2 and 5hmC and decreased 5mC levels. Considering that the TET2 gene.promoter contains response elements for transcription factors activated by cytokines, together to in vitro results suggest that changes in DNA hydroxymethylation, resulting from altered levels of TET2 are likely to be relevant in the Sjögren's syndrome etiopathogenesis.


Subject(s)
5-Methylcytosine/analogs & derivatives , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Methyl-CpG-Binding Protein 2/genetics , Proto-Oncogene Proteins/genetics , Salivary Glands, Minor/metabolism , Sjogren's Syndrome/genetics , 5-Methylcytosine/metabolism , Adult , Aged , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Cytokines/immunology , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Dioxygenases/immunology , Dioxygenases/metabolism , Epigenesis, Genetic , Female , Gene Expression , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/metabolism , Lip , Male , Methyl-CpG-Binding Protein 2/metabolism , Middle Aged , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , Mixed Function Oxygenases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Real-Time Polymerase Chain Reaction , Salivary Glands, Minor/cytology , Salivary Glands, Minor/immunology , Sjogren's Syndrome/immunology , Sjogren's Syndrome/metabolism , Young Adult , DNA Methyltransferase 3B
8.
Chembiochem ; 18(13): 1155-1171, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28423240

ABSTRACT

About 2-3 million years ago, Alu-mediated deletion of a critical exon in the CMAH gene became fixed in the hominin lineage ancestral to humans, possibly through a stepwise process of selection by pathogen targeting of the CMAH product (the sialic acid Neu5Gc), followed by reproductive isolation through female anti-Neu5Gc antibodies. Loss of CMAH has occurred independently in some other lineages, but is functionally intact in Old World primates, including our closest relatives, the chimpanzee. Although the biophysical and biochemical ramifications of losing tens of millions of Neu5Gc hydroxy groups at most cell surfaces remains poorly understood, we do know that there are multiscale effects functionally relevant to both sides of the host-pathogen interface. Hominin CMAH loss might also contribute to understanding human evolution, at the time when our ancestors were starting to use stone tools, increasing their consumption of meat, and possibly hunting. Comparisons with chimpanzees within ethical and practical limitations have revealed some consequences of human CMAH loss, but more has been learned by using a mouse model with a human-like Cmah inactivation. For example, such mice can develop antibodies against Neu5Gc that could affect inflammatory processes like cancer progression in the face of Neu5Gc metabolic incorporation from red meats, display a hyper-reactive immune system, a human-like tendency for delayed wound healing, late-onset hearing loss, insulin resistance, susceptibility to muscular dystrophy pathologies, and increased sensitivity to multiple human-adapted pathogens involving sialic acids. Further studies in such mice could provide a model for other human-specific processes and pathologies involving sialic acid biology that have yet to be explored.


Subject(s)
Genome , Hearing Loss/metabolism , Mixed Function Oxygenases/genetics , Muscular Dystrophies/metabolism , Neoplasms/metabolism , Neuraminic Acids/metabolism , Animals , Autoantibodies/biosynthesis , Biological Evolution , Disease Susceptibility , Gene Expression , Hearing Loss/genetics , Hearing Loss/immunology , Hearing Loss/pathology , Humans , Insulin Resistance , Mice , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/immunology , Muscular Dystrophies/genetics , Muscular Dystrophies/immunology , Muscular Dystrophies/pathology , Mutation , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neuraminic Acids/chemistry , Neuraminic Acids/immunology , Pan troglodytes
9.
Nat Chem Biol ; 11(9): 733-40, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258762

ABSTRACT

Distinct modifications fine-tune the activity of jasmonic acid (JA) in regulating plant growth and immunity. Hydroxylated JA (12OH-JA) promotes flower and tuber development but prevents induction of JA signaling, plant defense or both. However, biosynthesis of 12OH-JA has remained elusive. We report here an antibiotic biosynthesis monooxygenase (Abm) that converts endogenous free JA into 12OH-JA in the model rice blast fungus Magnaporthe oryzae. Such fungal 12OH-JA is secreted during host penetration and helps evade the defense response. Loss of Abm in M. oryzae led to accumulation of methyl JA (MeJA), which induces host defense and blocks invasive growth. Exogenously added 12OH-JA markedly attenuated abmΔ-induced immunity in rice. Notably, Abm itself is secreted after invasion and most likely converts plant JA into 12OH-JA to facilitate host colonization. This study sheds light on the chemical arms race during plant-pathogen interaction, reveals Abm as an antifungal target and outlines a synthetic strategy for transformation of a versatile small-molecule phytohormone.


Subject(s)
Cyclopentanes/metabolism , Fungal Proteins/immunology , Gene Expression Regulation, Fungal , Magnaporthe/genetics , Mixed Function Oxygenases/immunology , Oryza/immunology , Oxylipins/metabolism , Plant Growth Regulators/metabolism , Cyclopentanes/chemistry , Cyclopentanes/immunology , Fungal Proteins/chemistry , Fungal Proteins/genetics , Host-Pathogen Interactions/immunology , Hydroxylation , Magnaporthe/immunology , Magnaporthe/pathogenicity , Methylation , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Models, Molecular , Oryza/microbiology , Oxylipins/chemistry , Oxylipins/immunology , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Growth Regulators/chemistry , Plant Growth Regulators/immunology , Plant Immunity , Plant Leaves/immunology , Plant Leaves/microbiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction
10.
Exp Parasitol ; 176: 66-74, 2017 May.
Article in English | MEDLINE | ID: mdl-28327439

ABSTRACT

Leishmaniasis is a parasitic disease caused by the protozoan of the Leishmania genus. While no human vaccine is available, drugs such as pentavalent antimonials, pentamidine and amphotericin B are used for treat the patients. However, the high toxicity of these pharmaceutics, the emergence of parasite resistance and/or their high cost have showed to the urgent need of identify new targets to be employed in the improvement of the treatment against leishmaniasis. In a recent immunoproteomics approach performed in the Leishmania infantum species, 104 antigenic proteins were recognized by antibodies in sera of visceral leishmaniasis (VL) dogs. Some of them were later showed to be effective diagnostic markers and/or vaccine candidates against the disease. Between these proteins, 24 considered as hypothetical were identified in the promastigote and amastigote-like extracts of the parasites. The present study aimed to use bioinformatics tools to select new drug targets between these hypothetical proteins. Their cellular localization was predicted to be seven membrane proteins, as well as eight cytoplasmic, three nuclear, one mitochondrial and five proteins remained unclassified. Their functions were predicted as being two transport proteins, as well as five with metabolic activity, three as cell signaling and fourteen proteins remained unclassified. Ten hypothetical proteins were well-annotated and compared to their homology regarding to human proteins. Two proteins, a calpain-like and clavaminate synthase-like proteins were selected by using Docking analysis as being possible drug targets. In this sense, the present study showed the employ of new strategies to select possible drug candidates, according their localization and biological function in Leishmania parasites, aiming to treat against VL.


Subject(s)
Computational Biology/methods , Leishmania infantum/drug effects , Proteomics/methods , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Calpain/chemistry , Calpain/drug effects , Calpain/immunology , Drug Delivery Systems , Humans , Leishmania infantum/chemistry , Leishmania infantum/immunology , Leishmaniasis, Visceral/drug therapy , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/drug effects , Mixed Function Oxygenases/immunology , Models, Structural , Molecular Conformation , Protozoan Proteins/chemistry , Protozoan Proteins/drug effects , ROC Curve
11.
Glycobiology ; 26(6): 571-81, 2016 06.
Article in English | MEDLINE | ID: mdl-26935763

ABSTRACT

As a transplant surgeon, my interest in glycobiology began through my research into ABO-incompatible allotransplantation, and grew when my goal became overcoming the shortage of organs from deceased human donors by the transplantation of pig organs into patients with terminal organ failure (xenotransplantation/cross-species transplantation). The major target for human "natural" (preformed) anti-pig antibodies is galactose-α(1,3)-galactose (the "Gal" epitope), which is expressed on many pig cells, including the vascular endothelium. The binding of human IgM and IgG antibodies to Gal antigens initiates the process of hyperacute rejection, resulting in destruction of the pig graft within minutes or hours. This major barrier has been overcome by the production of pigs in which the gene for the enzyme α(1,3)-galactosyltransferase (GT) has been deleted by genetic engineering, resulting in GT knockout (GTKO) pigs. The two other known carbohydrate antigenic targets on pig cells for human anti-pig antibodies are (i) the product of the cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) gene, i.e., N-glycolylneuraminic acid, and (ii) the product of the ß1,4 N-acetylgalactosaminyltransferase gene, i.e., the Sd(a) antigen. Expression of these two has also been deleted in pigs. These genetic manipulations, together with others directed to overcoming primate complement and coagulation activation (the latter of which also relates to glycobiology) have contributed to the prolongation of pig graft survival in nonhuman primate recipients to many months rather than a few minutes. Clinical trials of the transplantation of pig cells are already underway and transplantation of pig organs may be expected within the relatively near future.


Subject(s)
Galactosyltransferases/genetics , Graft Rejection/prevention & control , Graft Survival , Mixed Function Oxygenases/genetics , Organ Transplantation/methods , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , Antigens/biosynthesis , Antigens/immunology , Galactosyltransferases/deficiency , Galactosyltransferases/immunology , Gene Deletion , Gene Expression , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Mixed Function Oxygenases/drug effects , Mixed Function Oxygenases/immunology , Papio , Swine , Transplantation, Homologous
12.
J Surg Res ; 200(2): 698-706, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26375504

ABSTRACT

BACKGROUND: Thrombocytopenia may represent a significant challenge to the clinical application of solid-organ xenotransplantation. When studied in a pig-to-primate model, consumptive coagulopathy has challenged renal xenografts. New strategies of genetic manipulation have altered porcine carbohydrate profiles to significantly reduce human antibody binding to pig cells. As this process continues to eliminate immunologic barriers to clinical xenotransplantation, the relationship between human platelets and pig organs must be considered. METHODS: Genetically modified pigs that were created by the CRISPR/Cas9 system with α-1,3-galactosyltransferase (GGTA1)(-/-) or GGTA1(-/-) cytidine monophosphate-N-acetylneuraminic acid hydroxylase(-/-) phenotype, as well as domestic pigs, were used in this study. Autologous porcine platelets were isolated from donor animal blood collection, and human platelets were obtained from a blood bank. Platelets were fluorescently labeled and in a single-pass model, human, or autologous platelets were perfused through porcine organs at a constant concentration and controlled temperature. Platelet uptake was measured by sampling venous output and measuring sample florescence against input florescence. In vitro study of the interaction between human platelets and porcine endothelial cells was accomplished by immunohistochemical stain and confocal microscopy. RESULTS: Differences between human and autologous platelet loss through the porcine kidney were not significant in any genetic background tested (WT P = 0.15, GGTA1(-/-)P = 0.12, GGTA1(-/-) cytidine monophosphate-N-acetylneuraminic acid hydroxylase(-/-)P = 0.25). The unmodified porcine liver consumed human platelets in a single-pass model of platelet perfusion in fewer than 10 min. WT suprahepatic inferior vena cava fluoresce reached a maximum of 76% of input fluoresce within the human platelet cohort and was significantly lower than the autologous platelet control cohort (P = 0.001). Confocal microscopic analysis did not demonstrate a significant association between human platelets and porcine renal endothelial cells compared with porcine liver endothelial positive controls. CONCLUSIONS: Our results suggest that in the absence of immunologic injury, human platelets respond in a variable fashion to organ-specific porcine endothelial surfaces. Human platelets are not removed from circulation by exposure to porcine renal endothelium but are removed by unmodified porcine hepatic endothelium. Kidneys possessing genetic modifications currently relevant to clinical xenotransplantation failed to consume human platelets in an isolated single-pass model. Human platelets did not exhibit significant binding to renal endothelial cells by in vitro assay.


Subject(s)
Animals, Genetically Modified , Blood Platelets/immunology , Kidney Transplantation/methods , Postoperative Complications/prevention & control , Sus scrofa/genetics , Thrombocytopenia/prevention & control , Transplantation, Heterologous/methods , Animals , Blood Platelets/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium/immunology , Endothelium/metabolism , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knockout Techniques , Humans , Kidney/immunology , Kidney/metabolism , Liver/immunology , Liver/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , Random Allocation , Sus scrofa/immunology , Swine , Thrombocytopenia/etiology
13.
Transplantation ; 105(2): 318-327, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32796494

ABSTRACT

BACKGROUND: Natural preformed anti-pig IgM/IgG antibodies in primates play an important role in xenograft rejection. As it is not clear how IgE and IgA engage in the immune system in xenotransplantation, we investigated natural preformed and elicited anti-pig IgE/IgA in naive primates and after xenotransplantation in nonhuman primates. METHODS: The binding of IgM/IgG/IgE/IgA antibodies to red blood cells (RBCs) from wild-type (WT), α1,3-galactosyltransferase gene-knockout (GTKO), and GTKO/cytidine monophospho-N-acetylneuraminic acid hydroxylase gene-knockout/ß-1,4 N-acetylgalactosaminyltransferase 2 gene-knockout (ie, triple-knockout pigs) pigs were measured by flow cytometry in naive human (n = 50) and baboon (n = 14) sera. Antibody binding to WT and GTKO pig RBCs (pRBCs) was also measured in the sera of baboons (nonsensitized n = 7, sensitized n = 2) and rhesus monkeys (nonsensitized n = 2, sensitized n = 11) following WT or GTKO pig organ/tissue xenotransplantation. Deposition of IgM/IgG/IgE/IgA in the grafts was detected by immunohistochemistry. RESULTS: The majority of humans had natural preformed IgM/IgG/IgE/IgA to WT and GTKO pRBCs. In contrast, IgM/IgG/IgE/IgA to triple-knockout pRBCs were present at lower levels and frequency (P < 0.01). Baboons also had IgM/IgG/IgE/IgA antibodies against WT pRBCs, but fewer to GTKO and triple-knockout (P < 0.01). After xenotransplantation into nonhuman primates, when IgM/IgG increased, IgE/IgA also increased, but to a lesser extent. In addition to IgM/IgG, IgE or IgA deposition was observed in rejected pig xenografts. CONCLUSIONS: Primates develop serum anti-pig IgE/IgA antibodies both naturally and during xenograft rejection. The pathophysiological role, if any, of anti-pig IgE/IgA antibodies remains unknown.


Subject(s)
Antibodies, Heterophile/blood , Erythrocytes/immunology , Graft Rejection/immunology , Immunoglobulin A/blood , Immunoglobulin E/blood , Transplantation, Heterologous/adverse effects , Animals , Animals, Genetically Modified , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Graft Rejection/blood , Graft Rejection/pathology , Humans , Macaca mulatta , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , Papio , Species Specificity , Sus scrofa/genetics , Sus scrofa/immunology
14.
Sci Rep ; 11(1): 13131, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162938

ABSTRACT

Xenotransplantation (cross-species transplantation) using genetically-engineered pig organs offers a potential solution to address persistent organ shortage. Current evaluation of porcine genetic modifications is to monitor the nonhuman primate immune response and survival after pig organ xenotransplantation. This measure is an essential step before clinical xenotransplantation trials, but it is time-consuming, costly, and inefficient with many variables. We developed an efficient approach to quickly examine human-to-pig xeno-immune responses in vitro. A porcine endothelial cell was characterized and immortalized for genetic modification. Five genes including GGTA1, CMAH, ß4galNT2, SLA-I α chain, and ß2-microglobulin that are responsible for the production of major xenoantigens (αGal, Neu5Gc, Sda, and SLA-I) were sequentially disrupted in immortalized porcine endothelial cells using CRISPR/Cas9 technology. The elimination of αGal, Neu5Gc, Sda, and SLA-I dramatically reduced the antigenicity of the porcine cells, though the cells still retained their ability to provoke human natural killer cell activation. In summary, evaluation of human immune responses to genetically modified porcine cells in vitro provides an efficient method to identify ideal combinations of genetic modifications for improving pig-to-human compatibility, which should accelerate the application of xenotransplantation to humans.


Subject(s)
Animals, Genetically Modified/immunology , Antigens, Heterophile/immunology , Endothelial Cells/immunology , Swine/immunology , Transplantation, Heterologous/methods , Animals , Antibodies, Heterophile/immunology , Antigen-Antibody Reactions , Antigens, Heterophile/genetics , CRISPR-Cas Systems , Cell Degranulation , Cell Line, Transformed , Cytokines/pharmacology , Endothelial Cells/drug effects , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knockout Techniques , Graft Rejection/immunology , Graft Rejection/prevention & control , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/immunology , Liver/cytology , Lymphocyte Activation , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
15.
J Exp Med ; 190(2): 169-76, 1999 Jul 19.
Article in English | MEDLINE | ID: mdl-10432280

ABSTRACT

Hepatitis C virus (HCV) is thought to be involved in the pathogenesis of autoimmune hepatitis (AIH) type 2, which is defined by the presence of type I antiliver kidney microsome autoantibodies directed mainly against cytochrome P450 (CYP)2D6 and by autoreactive liver infiltrating T cells. Virus-specific CD8(+) cytotoxic T lymphocytes (CTLs) that recognize infected cells and contribute to viral clearance and tissue injury during HCV infection could be involved in the induction of AIH. To explore whether the antiviral cellular immunity may turn against self-antigens, we characterized the primary CTL response against an HLA-A*0201-restricted HCV-derived epitope, i.e., HCV core 178-187, which shows sequence homology with human CYP2A6 and CYP2A7 8-17. To determine the relevance of these homologies for the pathogenesis of HCV-associated AIH, we used synthetic peptides to induce primary CTL responses in peripheral blood mononuclear cells of healthy blood donors and patients with chronic HCV infection. We found that the naive CTL repertoire of both groups contains cross-reactive CTLs inducible by the HCV peptide recognizing both CYP2A6 and CYP2A7 peptides as well as endogenously processed CYP2A6 protein. Importantly, we failed to induce CTLs with the CYP-derived peptides that showed a lower capacity to form stable complexes with the HLA-A2 molecule. These findings demonstrate the potential of HCV to induce autoreactive CD8(+) CTLs by molecular mimicry, possibly contributing to virus-associated autoimmunity.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/immunology , Hepacivirus/immunology , Molecular Mimicry , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Cross Reactions , Cytochrome P-450 CYP2A6 , Cytochrome P-450 Enzyme System/genetics , Epitopes/genetics , HLA-A2 Antigen , Hepacivirus/genetics , Hepatitis C/immunology , Hepatitis C Antigens/genetics , Humans , Liver/immunology , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , Sequence Homology, Amino Acid
16.
Exp Parasitol ; 124(4): 453-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19945457

ABSTRACT

Two genes from Trypanosoma brucei brucei are predicted to encode Fe(II)- and alpha-ketoglutarate-dependent enzymes related to fungal thymine 7-hydroxylase. Transcription of the thymine hydroxylase-like genes is up-regulated in the bloodstream form of the parasite over the insect form, whereas Western blot analysis indicates more cross-reactive protein in the latter life stage. The genes were cloned, the proteins purified from Escherichia coli, and both proteins were shown to bind Fe(II) and alpha-ketoglutarate, confirming proper folding. The isolated proteins were incubated with Fe(II)- and alpha-ketoglutarate plus thymine, thymidine, and other putative substrates, but no activity was detected. Furthermore, no thymine 7-hydroxylase activity was detected in extracts of procyclic or bloodstream form cells. Although the functions of these proteins remain unknown, we conclude they are unlikely to be involved in thymine salvage.


Subject(s)
Mixed Function Oxygenases/genetics , Protozoan Proteins/genetics , Trypanosoma brucei brucei/enzymology , Amino Acid Sequence , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/immunology , Blotting, Western , Escherichia coli/genetics , Gas Chromatography-Mass Spectrometry , Gene Expression Regulation, Enzymologic , Isoenzymes/biosynthesis , Isoenzymes/chemistry , Isoenzymes/genetics , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/immunology , Molecular Sequence Data , Polymerase Chain Reaction , Protozoan Proteins/biosynthesis , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Rabbits , Rhodotorula/enzymology , Rhodotorula/genetics , Spectrophotometry, Ultraviolet , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/immunology
17.
Oncol Rep ; 44(6): 2364-2372, 2020 12.
Article in English | MEDLINE | ID: mdl-33125119

ABSTRACT

Aspartate/asparagine ß­hydroxylase (AspH) is a type II transmembrane protein that catalyzes the post­translational hydroxylation of definite aspartyl and asparaginyl residues in epidermal growth factor­like domains of substrates. In the last few decades, accumulating evidence has indicated that AspH expression is upregulated in numerous types of human malignant cancer and is associated with poor survival and prognosis. The AspH protein aggregates on the surface of tumor cells, which contributes to inducing tumor cell migration, infiltration and metastasis. However, small­molecule inhibitors targeting hydroxylase activity can markedly block these processes, both in vitro and in vivo. Immunization of tumor­bearing mice with a phage vaccine fused with the AspH protein can substantially delay tumor growth and progression. Additionally, AspH antigen­specific CD4+ and CD8+ T cells were identified in the spleen of tumor­bearing mice. Therefore, these agents may be used as novel strategies for cancer treatment. The present review summarizes the current progress on the underlying mechanisms of AspH expression in cancer development.


Subject(s)
Antigens, Neoplasm/genetics , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/immunology , Membrane Proteins/genetics , Mixed Function Oxygenases/genetics , Muscle Proteins/genetics , Neoplasms/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/immunology , Furans/pharmacology , Furans/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/immunology , Mixed Function Oxygenases/metabolism , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/immunology , Muscle Proteins/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Sulfonic Acids/pharmacology , Sulfonic Acids/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Up-Regulation , Xenograft Model Antitumor Assays
18.
Front Immunol ; 10: 2396, 2019.
Article in English | MEDLINE | ID: mdl-31681287

ABSTRACT

The two major sialic acids described in mammalian cells are the N-glycolylneuraminic acid (Neu5Gc) and the N-acetylneuraminic acid (Neu5Ac). Neu5Gc synthesis starts from the N-acetylneuraminic acid (Neu5Ac) precursor modified by an hydroxylic group addition catalyzed by CMP-Neu5Ac hydroxylase enzyme (CMAH). In humans, CMAH was inactivated by a 92 bp deletion occurred 2-3 million years ago. Few other mammals do not synthetize Neu5Gc, however livestock species used for food production and as a source of biological materials for medical applications carry Neu5Gc. Trace amounts of Neu5Gc are up taken through the diet and incorporated into various tissues including epithelia and endothelia cells. Humans carry "natural," diet-induced Anti-Neu5Gc antibodies and when undertaking medical treatments or receiving transplants or devices that contain animal derived products they can cause immunological reaction affecting pharmacology, immune tolerance, and severe side effect like serum sickness disease (SSD). Neu5Gc null mice have been the main experimental model to study such phenotype. With the recent advances in genome editing, pigs and cattle KO for Neu5Gc have been generated always in association with the αGal KO. These large animals are normal and fertile and provide additional experimental models to study such mutation. Moreover, they will be the base for the development of new therapeutic applications like polyclonal IgG immunotherapy, Bioprosthetic Heart Valves, cells and tissues replacement.


Subject(s)
Mixed Function Oxygenases , Neuraminic Acids/immunology , Animals , Humans , Mice , Mice, Knockout , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylneuraminic Acid
19.
Front Immunol ; 10: 789, 2019.
Article in English | MEDLINE | ID: mdl-31134048

ABSTRACT

The glycocalyx of human cells differs from that of many other mammals by the lack of the sialic acid N-glycolylneuraminic acid (Neu5Gc) and increased abundance of its precursor N-acetylneuraminic acid (Neu5Ac). Most humans also have circulating antibodies specifically targeting the non-human sialic acid Neu5Gc. Recently, several additional mammalian species have been found to also lack Neu5Gc. In all cases, loss-of-function mutations in the gene encoding the sialic acid-modifying enzyme CMAH are responsible for the drastic change in these species. Unlike other glycan antigens, Neu5Gc apparently cannot be produced by microbes, raising the question about the origin of these antibodies in humans. Dietary exposure and presentation on bacteria coating themselves with Neu5Gc from the diet are distinct possibilities. However, the majority of the non-human species that lack Neu5Gc do not consume diets rich in Neu5Gc, making it unlikely that they will have been immunized against this sialic acid. A notable exception are mustelids (ferrets, martens and their relatives) known for preying on various small mammal species rich in Neu5Gc. No studies exist on levels of anti-Neu5Gc antibodies in non-human species. Evolutionary scenarios for the repeated, independent fixation of CMAH loss-of-function mutations at various time points in the past include strong selection by parasites, especially enveloped viruses, stochastic effects of genetic drift, and directional selection via female immunity to paternal Neu5Gc. Convergent evolution of losses of the vertebrate-specific self-glycan Neu5Gc are puzzling and may represent a prominent way in which glycans become agents of evolutionary change in their own right. Such change may include the reconfiguration of innate immune lectins that use self-sialic acids as recognition patterns.


Subject(s)
Antibodies/immunology , Evolution, Molecular , Glycocalyx , Loss of Function Mutation , Mixed Function Oxygenases/deficiency , Neuraminic Acids , Animals , Female , Glycocalyx/genetics , Glycocalyx/immunology , Humans , Male , Mixed Function Oxygenases/immunology
20.
Front Immunol ; 10: 807, 2019.
Article in English | MEDLINE | ID: mdl-31057542

ABSTRACT

The description of "serum sickness" more than a century ago in humans transfused with animal sera eventually led to identification of a class of human antibodies directed against glycans terminating in the common mammalian sialic acid N-Glycolylneuraminic acid (Neu5Gc), hereafter called "Neu5Gc-glycans." The detection of such glycans in malignant and fetal human tissues initially raised the possibility that it was an oncofetal antigen. However, "serum sickness" antibodies were also noted in various human disease states. These findings spurred further research on Neu5Gc, and the discovery that it is not synthesized in the human body due to a human-lineage specific genetic mutation in the enzyme CMAH. However, with more sensitive techniques Neu5Gc-glycans were detected in smaller quantities on certain human cell types, particularly epithelia and endothelia. The likely explanation is metabolic incorporation of Neu5Gc from dietary sources, especially red meat of mammalian origin. This incorporated Neu5Gc on glycans appears to be the first example of a "xeno-autoantigen," against which varying levels of "xeno-autoantibodies" are present in all humans. The resulting chronic inflammation or "xenosialitis" may have important implications in human health and disease, especially in conditions known to be aggravated by consumption of red meat. In this review, we will cover the early history of the discovery of "serum sickness" antibodies, the subsequent recognition that they were partly directed against Neu5Gc-glycans, the discovery of the genetic defect eliminating Neu5Gc production in humans, and the later recognition that this was not an oncofetal antigen but the first example of a "xeno-autoantigen." Further, we will present comments about implications for disease risks associated with red meat consumption such as cancer and atherosclerosis. We will also mention the potential utility of these anti-Neu5Gc-glycan antibodies in cancer immunotherapy and provide some suggestions and perspectives for the future. Other reviews in this special issue cover many other aspects of this unusual pathological process, for which there appears to be no other described precedent.


Subject(s)
Autoantibodies/immunology , Immune System Diseases , Mixed Function Oxygenases , N-Acetylneuraminic Acid/immunology , Sialic Acids/immunology , Animals , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylneuraminic Acid/genetics , Sialic Acids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL