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










Publication year range
1.
Nat Commun ; 15(1): 1405, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360940

ABSTRACT

Mechanical force contributes to perforin pore formation at immune synapses, thus facilitating the cytotoxic T lymphocytes (CTL)-mediated killing of tumor cells in a unidirectional fashion. How such mechanical cues affect CTL evasion of perforin-mediated autolysis remains unclear. Here we show that activated CTLs use their softness to evade perforin-mediated autolysis, which, however, is shared by T leukemic cells to evade CTL killing. Downregulation of filamin A is identified to induce softness via ZAP70-mediated YAP Y357 phosphorylation and activation. Despite the requirements of YAP in both cell types for softness induction, CTLs are more resistant to YAP inhibitors than malignant T cells, potentially due to the higher expression of the drug-resistant transporter, MDR1, in CTLs. As a result, moderate inhibition of YAP stiffens malignant T cells but spares CTLs, thus allowing CTLs to cytolyze malignant cells without autolysis. Our findings thus hint a mechanical force-based immunotherapeutic strategy against T cell leukemia.


Subject(s)
Cytotoxicity, Immunologic , T-Lymphocytes, Cytotoxic , Perforin/genetics , Perforin/metabolism , Pore Forming Cytotoxic Proteins/genetics
3.
Nat Immunol ; 24(12): 2042-2052, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37919525

ABSTRACT

Tumor-derived factors are thought to regulate thrombocytosis and erythrocytopenia in individuals with cancer; however, such factors have not yet been identified. Here we show that tumor cell-released kynurenine (Kyn) biases megakaryocytic-erythroid progenitor cell (MEP) differentiation into megakaryocytes in individuals with cancer by activating the aryl hydrocarbon receptor-Runt-related transcription factor 1 (AhR-RUNX1) axis. During tumor growth, large amounts of Kyn from tumor cells are released into the periphery, where they are taken up by MEPs via the transporter SLC7A8. In the cytosol, Kyn binds to and activates AhR, leading to its translocation into the nucleus where AhR transactivates RUNX1, thus regulating MEP differentiation into megakaryocytes. In addition, activated AhR upregulates SLC7A8 in MEPs to induce positive feedback. Importantly, Kyn-AhR-RUNX1-regulated MEP differentiation was demonstrated in both humanized mice and individuals with cancer, providing potential strategies for the prevention of thrombocytosis and erythrocytopenia.


Subject(s)
Neoplasms , Thrombocytosis , Animals , Mice , Kynurenine/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Megakaryocytes/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Erythroid Precursor Cells/metabolism , Cell Differentiation/physiology , Neoplasms/metabolism , Thrombocytosis/metabolism , Bias
4.
Cell Death Differ ; 29(1): 246-261, 2022 01.
Article in English | MEDLINE | ID: mdl-34453118

ABSTRACT

T cell-driven diseases account for considerable morbidity and disability globally and there is an urgent need for new targeted therapies. Both cancer cells and activated T cells have an altered redox balance, and up-regulate the DNA repair protein MTH1 that sanitizes the oxidized nucleotide pool to avoid DNA damage and cell death. Herein we suggest that the up-regulation of MTH1 in activated T cells correlates with their redox status, but occurs before the ROS levels increase, challenging the established conception of MTH1 increasing as a direct response to an increased ROS status. We also propose a heterogeneity in MTH1 levels among activated T cells, where a smaller subset of activated T cells does not up-regulate MTH1 despite activation and proliferation. The study suggests that the vast majority of activated T cells have high MTH1 levels and are sensitive to the MTH1 inhibitor TH1579 (Karonudib) via induction of DNA damage and cell cycle arrest. TH1579 further drives the surviving cells to the MTH1low phenotype with altered redox status. TH1579 does not affect resting T cells, as opposed to the established immunosuppressor Azathioprine, and no sensitivity among other major immune cell types regarding their function can be observed. Finally, we demonstrate a therapeutic effect in a murine model of experimental autoimmune encephalomyelitis. In conclusion, we show proof of concept of the existence of MTH1high and MTH1low activated T cells, and that MTH1 inhibition by TH1579 selectively suppresses pro-inflammatory activated T cells. Thus, MTH1 inhibition by TH1579 may serve as a novel treatment option against autoreactive T cells in autoimmune diseases, such as multiple sclerosis.


Subject(s)
DNA Repair Enzymes , Phosphoric Monoester Hydrolases , Animals , DNA Damage , DNA Repair Enzymes/metabolism , Lymphocyte Count , Mice , Phosphoric Monoester Hydrolases/genetics , T-Lymphocytes/metabolism
5.
Cancer Res ; 81(2): 476-488, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33168645

ABSTRACT

Biomechanics is a fundamental feature of a cell. However, the manner by which actomysin tension affects tumor immune evasion remains unclear. Here we show that although cytotoxic T lymphocytes (CTL) can effectively destroy stiff differentiated tumor cells, they fail to kill soft tumor-repopulating cells (TRC). TRC softness prevented membrane pore formation caused by CTL-released perforin. Perforin interacting with nonmuscle myosin heavy-chain 9 transmitted forces to less F-actins in soft TRC, thus generating an inadequate contractile force for perforin pore formation. Stiffening TRC allowed perforin the ability to drill through the membrane, leading to CTL-mediated killing of TRC. Importantly, overcoming mechanical softness in human TRC also enhanced TRC cell death caused by human CTL, potentiating a mechanics-based immunotherapeutic strategy. These findings reveal a mechanics-mediated tumor immune evasion, thus potentially providing an alternative approach for tumor immunotherapy. SIGNIFICANCE: Tumor-repopulating cells evade CD8+ cytolytic T-cell killing through a mechanical softness mechanism, underlying the impediment of perforin pore formation at the immune synapse site.


Subject(s)
Colonic Neoplasms/pathology , Cytotoxicity, Immunologic/immunology , Melanoma/pathology , Perforin/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Apoptosis , Cell Proliferation , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Female , Humans , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cancer Immunol Res ; 6(9): 1057-1068, 2018 09.
Article in English | MEDLINE | ID: mdl-30018046

ABSTRACT

Tumor cell-derived microparticles (T-MP) contain tumor antigen profiles as well as innate signals, endowing them with vaccine potential; however, the precise mechanism by which DCs present T-MP antigens to T cells remains unclear. Here, we show that T-MPs activate a lysosomal pathway that is required for DCs presenting tumor antigens of T-MPs. DCs endocytose T-MPs to lysosomes, where T-MPs increase lysosomal pH from 5.0 to a peak of 8.5 via NOX2-catalyzed reactive oxygen species (ROS) production. This increased pH, coupled with T-MP-driven lysosomal centripetal migration, promotes the formation of MHC class I-tumor antigen peptide complexes. Concurrently, endocytosis of T-MPs results in the upregulation of CD80 and CD86. T-MP-increased ROS activate lysosomal Ca2+ channel Mcoln2, leading to Ca2+ release. Released Ca2+ activates transcription factor EB (TFEB), a lysosomal master regulator that directly binds to CD80 and CD86 promoters, promoting gene expression. These findings elucidate a pathway through which DCs efficiently present tumor antigen from T-MPs to CD8+ T cells, potentiating T-MPs as a novel tumor cell-free vaccine with clinical applications. Cancer Immunol Res; 6(9); 1057-68. ©2018 AACR.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell-Derived Microparticles/immunology , Dendritic Cells/immunology , Animals , B7-1 Antigen/genetics , B7-2 Antigen/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cancer Vaccines/immunology , Cell Differentiation , Cells, Cultured , Endocytosis/immunology , Female , Histocompatibility Antigens Class I/immunology , Lysosomes/physiology , Melanoma, Experimental , Mice , Mice, Inbred C57BL
7.
Nat Commun ; 9(1): 1808, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717137

ABSTRACT

In the originally published version of this Article, images in Fig. 5n were inadvertently replaced with duplicates of images in Fig. 5o during the production process. This has now been corrected in both the PDF and HTML versions of the Article.

8.
Cancer Res ; 78(14): 3926-3937, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29764867

ABSTRACT

Dormancy is recognized as a critical biological event for tumorigenic cells surviving in an extremely harsh environment. Understanding the molecular process of dormancy can unlock novel approaches to tackle cancers. We recently reported that stem-like tumor-repopulating cells (TRC) sense mechanical signals and rapidly proliferate in a 90 Pa soft fibrin matrix. Here, we show that a stiff mechanical environment induces TRC dormancy via an epigenetic program initiated by translocation of Cdc42, a cytosolic regulator of mechanotransduction, into the nucleus, where it promotes transcription of hydroxymethylating enzyme Tet2. Tet2 epigenetically activated cell-cycle-inhibiting genes p21 and p27 to induce dormancy, but also caused downregulation of integrin ß3 to maintain dormancy. This stiffness-mediated dormancy was recapitulated in mouse models for both murine and primary human melanoma TRCs. These data identify an epigenetic program directed by mechanics, which drives highly tumorigenic TRCs to enter dormancy in a stiff mechanical environment.Significance: A mechanics-directed epigenetic program enables tumor-repopulating cells to enter dormancy in a stiff mechanical environment. Cancer Res; 78(14); 3926-37. ©2018 AACR.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic/physiology , Fibrin/metabolism , Proto-Oncogene Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Dioxygenases , Down-Regulation/physiology , Epigenomics/methods , Female , Hep G2 Cells , Humans , Mechanotransduction, Cellular/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID
9.
Nat Commun ; 9(1): 873, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491374

ABSTRACT

Resetting tumor-associated macrophages (TAMs) is a promising strategy to ameliorate the immunosuppressive tumor microenvironment and improve innate and adaptive antitumor immunity. Here we show that chloroquine (CQ), a proven anti-malarial drug, can function as an antitumor immune modulator that switches TAMs from M2 to tumor-killing M1 phenotype. Mechanistically, CQ increases macrophage lysosomal pH, causing Ca2+ release via the lysosomal Ca2+ channel mucolipin-1 (Mcoln1), which induces the activation of p38 and NF-κB, thus polarizing TAMs to M1 phenotype. In parallel, the released Ca2+ activates transcription factor EB (TFEB), which reprograms the metabolism of TAMs from oxidative phosphorylation to glycolysis. As a result, CQ-reset macrophages ameliorate tumor immune microenvironment by decreasing immunosuppressive infiltration of myeloid-derived suppressor cells and Treg cells, thus enhancing antitumor T-cell immunity. These data illuminate a previously unrecognized antitumor mechanism of CQ, suggesting a potential new macrophage-based tumor immunotherapeutic modality.


Subject(s)
Antineoplastic Agents/pharmacology , Chloroquine/pharmacology , Immunotherapy/methods , Macrophages/cytology , Macrophages/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Cell Line, Tumor , Female , Glycolysis/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , NF-kappa B/metabolism , RAW 264.7 Cells , Transient Receptor Potential Channels/metabolism , Tumor Microenvironment/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Cancer Cell ; 33(3): 480-494.e7, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29533786

ABSTRACT

Despite the clinical successes fostered by immune checkpoint inhibitors, mechanisms underlying PD-1 upregulation in tumor-infiltrating T cells remain an enigma. Here, we show that tumor-repopulating cells (TRCs) drive PD-1 upregulation in CD8+ T cells through a transcellular kynurenine (Kyn)-aryl hydrocarbon receptor (AhR) pathway. Interferon-γ produced by CD8+ T cells stimulates release of high levels of Kyn produced by TRCs, which is transferred into adjacent CD8+ T cells via the transporters SLC7A8 and PAT4. Kyn induces and activates AhR and thereby upregulates PD-1 expression. This Kyn-AhR pathway is confirmed in both tumor-bearing mice and cancer patients and its blockade enhances antitumor adoptive T cell therapy efficacy. Thus, we uncovered a mechanism of PD-1 upregulation with potential tumor immunotherapeutic applications.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Kynurenine/pharmacology , Programmed Cell Death 1 Receptor/drug effects , Receptors, Aryl Hydrocarbon/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Interferon-gamma/immunology , Mice , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
11.
J Clin Invest ; 128(3): 1057-1073, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29431732

ABSTRACT

Dynamic interaction with the immune system profoundly regulates tumor cell dormancy. However, it is unclear how immunological cues trigger cancer cell-intrinsic signaling pathways for entering into dormancy. Here, we show that IFN-ß treatment induced tumor-repopulating cells (TRC) to enter dormancy through an indolamine 2,3-dioxygenase/kynurenine/aryl hydrocarbon receptor/p27-dependent (IDO/Kyn/AhR/p27-dependent) pathway. Strategies to block this metabolic circuitry did not relieve dormancy, but led to apoptosis of dormant TRCs in murine and human melanoma models. Specifically, blocking AhR redirected IFN-ß signaling to STAT3 phosphorylation through both tyrosine and serine sites, which subsequently facilitated STAT3 nuclear translocation and subsequent binding to the p53 promoter in the nucleus. Upregulation of p53 in turn disrupted the pentose phosphate pathway, leading to excessive ROS production and dormant TRC death. Additionally, in melanoma patients, high expression of IFN-ß correlated with tumor cell dormancy. Identification of this mechanism for controlling TRC dormancy by IFN-ß provides deeper insights into cancer-immune interaction and potential new cancer immunotherapeutic modalities.


Subject(s)
Interferon-beta/pharmacology , Neoplastic Stem Cells/cytology , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immune System , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , MCF-7 Cells , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Serine/chemistry , Signal Transduction , Tyrosine/chemistry
12.
Clin Immunol ; 166-167: 27-37, 2016 05.
Article in English | MEDLINE | ID: mdl-27102765

ABSTRACT

OBJECTIVES: Phosphorylcholine (PC) and malondialdehyde (MDA) are generated during lipid peroxidation and form adducts with proteins as albumin as studied herein. Atherosclerosis and cardiovascular disease (CVD) are increased in systemic lupus erythematosus (SLE). We here investigate the role and regulation of IgM antibodies against PC (anti-PC) and MDA (anti-MDA). METHODS: IgM anti-PC and anti-MDA in SLE patients (n=114) were compared with age- and sex-matched population-based controls (n=108). Common carotid intima-media thickness (IMT) and plaque occurrence were determined by B-mode ultrasound. Plaques were graded according to echogenicity (potentially vulnerability). Production of IgM anti-PC and anti-MDA by B cells was determined by ELISA and ELISPOT. The effect of anti-PC and anti-MDA on macrophage uptake of apoptotic cells and oxidative stress was studied by flow cytometry. RESULTS: Above 66rd percentile together, IgM anti-PC and anti-MDA were striking protection markers for plaque prevalence and echolucency in SLE (OR: 0.08, CI: 0.01-0.46 and OR: 0.10, CI: 0.01-0.82), respectively, and risk markers for plaque prevalence when below 33rd percentile: OR: 3.79, CI: (1.10-13.00). In vitro, IgM anti-PC and anti-MDA were much higher when B cells were co-cultured with CD3 T cells. Anti-HLA-, anti-CD40 antibody or CD40 silencing abolished these effects. Uptake of apoptotic cells was increased by IgM anti-PC and anti-MDA. MDA induced increased oxidative stress, which was inhibited by IgM anti-MDA. CONCLUSIONS: Unexpectedly, both IgM anti-MDA and IgM anti-PC are T-cell dependent and especially together, are strong protection markers for atherosclerosis in SLE. Underlying mechanisms include increased phagocytosis of apoptotic cells and decrease of oxidative stress.


Subject(s)
Atherosclerosis/immunology , Immunoglobulin M/immunology , Lupus Erythematosus, Systemic/immunology , Malondialdehyde/immunology , Phosphorylcholine/immunology , Adult , Apoptosis/immunology , Atherosclerosis/complications , Atherosclerosis/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , Carotid Intima-Media Thickness , Cells, Cultured , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Humans , Immunoglobulin M/metabolism , Jurkat Cells , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/metabolism , Male , Malondialdehyde/metabolism , Middle Aged , Oxidative Stress/immunology , Phagocytosis/immunology , Phosphorylcholine/metabolism , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Ultrasonography
13.
Arterioscler Thromb Vasc Biol ; 35(1): 197-205, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25395618

ABSTRACT

OBJECTIVE: Atherosclerosis is an inflammatory disease, where activated immunocompetent cells, including dendritic cells (DCs) and T cells are abundant in plaques. Low-density lipoprotein modified either by oxidation (oxLDL) or by human group X-secreted phospholipase A2 (LDLx) and heat shock proteins (HSP), especially HSP60 and 90, have been implicated in atherosclerosis. We previously reported that Annexin A5 inhibits inflammatory effects of phospholipids, decreases vascular inflammation and improves vascular function in apolipoprotein E(-/-) mice. Here, we focus on the LDLx effects on human DCs and T cells. APPROACH AND RESULTS: Human DCs were differentiated from peripheral blood monocytes, stimulated by oxLDL or LDLx. Naive autologous T cells were cocultured with pretreated DCs. oxLDL and LDLx, in contrast to LDL, induced DC-activation and T-cell proliferation. T cells exposed to LDLx-treated DCs produced interferon-γ, interleukin (IL)-17 but not IL-4 and IL-10. Annexin A5 abrogated LDLx effects on DCs and T cells and increased production of transforming growth factor-ß and IL-10. Furthermore, IL-10 producing T cells suppressed primary T-cell activation via soluble IL-10, transforming growth factor-ß, and cell-cell contact. Lentiviral-mediated shRNA knock-down HSP60 and 90 in DCs attenuated the effect of LDLx on DCs and subsequent T-cell proliferation. Experiments on DC and T cells derived from carotid atherosclerotic plaques gave similar results. CONCLUSIONS: Our data show that modified forms of LDL such as LDLx but not native LDL activate human T cells through DCs. HSP60 and 90 contribute to such T-cell activation. Annexin A5 promotes induction of regulatory T cells and is potentially interesting as a therapeutic agent.


Subject(s)
Annexin A5/metabolism , Cell Communication , Chaperonin 60/metabolism , Dendritic Cells/metabolism , HSP90 Heat-Shock Proteins/metabolism , Lipoproteins, LDL/metabolism , Lymphocyte Activation , Mitochondrial Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Carotid Artery Diseases/immunology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chaperonin 60/genetics , Coculture Techniques , Cytokines/metabolism , Dendritic Cells/immunology , Group X Phospholipases A2/metabolism , HSP90 Heat-Shock Proteins/genetics , Humans , Inflammation Mediators/metabolism , Mitochondrial Proteins/genetics , Plaque, Atherosclerotic , RNA Interference , Signal Transduction , T-Lymphocyte Subsets/immunology , Transfection
14.
J Immunol ; 192(10): 4551-9, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24729615

ABSTRACT

Phosphorylcholine (PC) is a classic T-independent Ag that is exposed on apoptotic cells, oxidized phospholipids, and bacterial polysaccharides. Experimental as well as epidemiological studies have over the past decade implicated Abs against PC (anti-PC) as anti-inflammatory and a strong protective factor in cardiovascular disease. Although clinically important, little is known about the development of anti-PC in humans. This study was conceived to dissect the human anti-PC repertoire and generate human mAbs. We designed a PC-specific probe to identify, isolate, and characterize PC-reactive B cells from 10 healthy individuals. The donors had all mounted somatically mutated Abs toward PC using a broad variety of Ig genes. PC-reactive B cells were primarily found in the IgM(+) memory subset, although significant numbers also were detected among naive, IgG(+), and CD27(+)CD43(+) B cells. Abs from these subsets were clonally related, suggesting a common origin. mAbs derived from the same donors exhibited equivalent or higher affinity for PC than the well-characterized murine T-15 clone. These results provide novel insights into the cellular and molecular ontogeny of atheroprotective PC Abs, thereby offering new opportunities for Ab-based therapeutic interventions.


Subject(s)
Antibodies, Antiphospholipid/immunology , B-Lymphocyte Subsets/immunology , Immunoglobulin M/immunology , Immunologic Memory/physiology , Phosphorylcholine/immunology , Adult , Animals , B-Lymphocyte Subsets/cytology , Female , Humans , Immunoglobulin G/immunology , Male , Mice
15.
J Mol Biol ; 417(4): 279-93, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22330145

ABSTRACT

The enzyme aldos-2-ulose dehydratase/isomerase (AUDH) participates in carbohydrate secondary metabolism, catalyzing the conversion of glucosone and 1,5-d-anhydrofructose to the secondary metabolites cortalcerone and microthecin, respectively. AUDH is a homo-dimeric enzyme with subunits of 900 amino acids. The subunit consists of a seven-bladed ß-propeller domain, two cupin folds and a C-terminal lectin domain. AUDH contains a structural Zn(2+) and Mg(2+) located in loop regions and two zinc ions at the bottom of two putative active-site clefts in the propeller and the cupin domain, respectively. Catalysis is dependent on these two zinc ions, as their specific removal led to loss of enzymatic activity. The structure of the Zn(2)(+)-depleted enzyme is very similar to that of native AUDH, and structural changes upon metal removal as the cause for the catalytic deficiencies can be excluded. The complex with the reaction intermediate ascopyrone M shows binding of this compound at two different sites, with direct coordination to Zn(2+) in the propeller domain and as second sphere ligand of the metal ion in the cupin domain. These observations suggest that the two reactions of AUDH might be catalyzed in two different active sites, about 60 Å apart. The dehydration reaction most likely follows an elimination mechanism, where Zn(2+) acts as a Lewis acid polarizing the C2 keto group of 1,5-d-anhydrofructose. Abstraction of the proton at the C3 carbon atom and protonation of the leaving group, the C4 hydroxyl moiety, could potentially be catalyzed by the side chain of the suitably positioned residue His155.


Subject(s)
Fructose/analogs & derivatives , Hydro-Lyases/chemistry , Phanerochaete/enzymology , Zinc/chemistry , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Fructose/chemistry , Ketoses/chemistry , Molecular Sequence Data , Protein Structure, Tertiary
16.
Results Immunol ; 2: 13-8, 2012.
Article in English | MEDLINE | ID: mdl-24371562

ABSTRACT

BACKGROUND: We have reported that anti-phosphorylcholine (anti-PC) IgM is a protection marker for human cardiovascular disease (CVD) and atherosclerosis. We here investigate the anti-PC autoantibodies in a well-defined cohort with regard to idiotype, atherosclerosis progression and mechanisms for its protective action. METHODS: Serum levels and binding specificities of different anti-PC isotypes were determined in 226 hypertensive individuals enrolled in European Lacidipine Study on Atherosclerosis using ELISA. The mean of the maximum Intima-Media Thicknesses (IMT) in the far walls of common carotids and bifurcations was assessed at the time of inclusion, and four years afterwards. Apoptosis in immune cells was induced with lysophosphatidylcholine (LPC) and quantified using the MTT-assay. RESULTS: Anti-PC IgM, IgA and IgG1 (but not IgG2) was negatively associated with IMT-progression. Combining anti-PC IgM with data on antibodies against oxidized- and malondialdehyde-modified LDL further strengthened this association. At very high levels, anti-PC IgM exhibited a striking negative association with atherosclerosis progression (OR 0.05; CI 0.006-0.40). Analysis of serum samples taken four years apart in study participants affirmed the stability of anti-PC IgM titers over time. Examination of fine specificities revealed that the protective isotypes (IgM, IgA and IgG1) are of the Group I idiotype whereas the non-protective IgG2 subclass was Group II. Anti-PC IgM inhibited LPC-induced cell death of immune cells. CONCLUSION: Group I anti-PC antibodies, particularly of the IgM class, are independent protection markers for atherosclerosis progression. One potential mechanism of action is inhibition of LPC-induced cell cytotoxicity.

17.
Clin Exp Rheumatol ; 29(6): 942-50, 2011.
Article in English | MEDLINE | ID: mdl-22153361

ABSTRACT

OBJECTIVES: To examine how treatment of rheumatoid arthritis (RA) with anti-tumour necrosis factor alpha antagonists (anti-TNF) and B-cell targeting rituximab influences novel markers of atherosclerosis and inflammation, such as atheroprotective natural IgM antibodies against phosphorylcholine (anti-PC), oxidised low-density lipoprotein (oxLDL) and apolipoproteins. METHODS: In a prospective study 215 patients with RA were enrolled of whom 85.6% were seropositive, aged 57.9 ± 12.4 years, with mean disease duration 8.5 (5-15) years. 162 patients were treated with anti-TNF and 53 with rituximab for one year. The patients were assessed and blood sampled at 0, 3, 6 and 12 months. IgM anti-PC and oxLDL were determined by ELISA and apolipoproteins by immunoturbidimetry. RESULTS: IgM anti-PC increased by 26% during anti-TNF treatment, p<0.001, while decreased by 14% on rituximab, p=0.023, after 12 months of treatment. Patients in remission after 12 months, DAS28<2.6, had higher baseline anti-PC levels compared with those not in remission in both anti-TNF, p=0.007, and rituximab-treated subjects, p=0.041. In both treatment groups, levels of oxLDL increased temporarily at three months but apoA1 improved throughout the study. This effect was inversely correlated with changes in disease activity. The apoB and apoB/apoA1-ratio remained stable throughout the whole study period. CONCLUSIONS: Anti-TNF treatment demonstrated a favourable long-term effect on anti-PC levels. Low levels of IgM anti-PC may identify immune-deficient state and predict inferior therapy response. Biological therapies increased the level of the anti-atherogenic lipid apoA1. The impact of these effects on future CVD events deserves further studies.


Subject(s)
Antirheumatic Agents/therapeutic use , Apolipoproteins/blood , Arthritis, Rheumatoid/therapy , Atherosclerosis/prevention & control , Immunologic Factors/therapeutic use , Phosphorylcholine/immunology , Adalimumab , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Arthritis, Rheumatoid/blood , Atherosclerosis/blood , Biomarkers/metabolism , Cholesterol, LDL/blood , Etanercept , Female , Humans , Immunoglobulin G/therapeutic use , Infliximab , Lipoproteins, LDL/blood , Male , Middle Aged , Prospective Studies , Receptors, Tumor Necrosis Factor/therapeutic use , Rituximab , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
18.
Planta Med ; 76(15): 1635-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20645241

ABSTRACT

1,5-Anhydro-D-fructose (AF) was first found in fungi and red algae. It is produced by the degradation of glycogen, starch and maltosaccharides with α-1,4-glucan lyase (EC 4.2.2.13). In vivo, AF is metabolized to 1,5-anhydro-D-glucitol (AG), ascopyrone P (APP), microthecin and other derivatives via the anhydrofructose pathway. The genes coding for the enzymes in this pathway have been cloned, enabling the large-scale production of AF and related products in a cell-free reactor. The possible applications of these products in medicine have been evaluated using both in vitro and in vivo systems. Thus AF is a useful anticariogenic agent as it inhibits the growth of the oral pathogen Streptococcus mutans, impairing the production of plaque-forming polysaccharides and lactic acid. AF also shows anti-inflammatory and anticancer effects. AG is used as a diabetic marker for glycemic control. AG also stimulates insulin secretion in insulinoma cell lines. in vivo, APP has been shown to lengthen the life span of cancer-afflicted mice. It interferes with tumor growth and metastasis by its cidal effects on fast multiplying cells. Microthecin inhibits the growth of the human pathogen Pseudomonas aeruginosa PAO1, particularly under anaerobic conditions. The pharmaceutical usefulness of the other AF metabolites 1,5-anhydro-D-mannitol,1-deoxymannojirimycin, haliclonol, 5-epipentenomycin I, bissetone, palythazine, isopalythazine, and clavulazine remains to be investigated. In this review AF and its metabolites as the bioactive natural products for their pharmaceutical potentials are discussed.


Subject(s)
Fructose/analogs & derivatives , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Antioxidants/metabolism , Antioxidants/pharmacology , Carbohydrate Metabolism , Dental Caries/prevention & control , Fructose/chemistry , Fructose/metabolism , Fructose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Ketoses/metabolism , Ketoses/pharmacology , Mice , Protective Agents/chemistry , Protective Agents/metabolism , Protective Agents/pharmacology , Rats
19.
Stroke ; 41(4): 607-12, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20150554

ABSTRACT

BACKGROUND AND PURPOSE: Natural immunoglobulin M antibodies specific for phosphorylcholine (anti-PC) have been implicated in atherosclerosis. We have shown previously that high levels of anti-PC predict a slower progression of atherosclerosis in humans and that low levels of anti-PC are associated with higher risk for cardiovascular disease. Here we determine the association between anti-PC and the incidence of stroke. METHODS: Using a nested case control study design, we examined 227 incident cases (125 men and 102 women) of first-time stroke and 455 age- and sex-matched controls identified during a 13-year time period (1985 to 1999) within the population-based cohorts of the Västerbotten Intervention Project (VIP) and the World Health Organization Monitoring Trends and Determinants in Cardiovascular Disease (WHO MONICA) project in Northern Sweden. Odds ratios of stroke with 95% CIs with adjustments for age, gender, smoking, serum cholesterol, diabetes, body mass index, and blood pressure were determined. Anti-PC levels were measured using ELISA. RESULTS: A significant association between low levels of anti-PC at baseline and incident stroke was seen for the whole group of anti-PC levels below the 30th percentile (multivariately adjusted odds ratio, 1.62; CI, 1.11 to 2.35). Analyses of gender-specific associations indicated fairly strong associations for females, especially at the lowest 30th percentile (multivariately adjusted odds ratio, 2.65; CI, 1.41 to 4.95). No associations were noted for men. CONCLUSION: Low anti-PC is a novel independent risk marker for development of stroke. Measurements of anti-PC could be used to identify immunodeficient subjects at an increased risk for stroke. The possibility that such subjects might be targets for novel modes of treatment such as immunotherapies deserves further investigation.


Subject(s)
Antibodies/blood , Phosphorylcholine/immunology , Stroke , Adult , Aged , Atherosclerosis/blood , Atherosclerosis/immunology , Case-Control Studies , Female , Humans , Immunoglobulin M/blood , Immunoglobulin M/immunology , Male , Middle Aged , Odds Ratio , Risk Factors , Stroke/blood , Stroke/epidemiology , Stroke/immunology , Sweden/epidemiology
20.
Biochim Biophys Acta ; 1760(9): 1314-22, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16822618

ABSTRACT

Anhydrofructose (AF) pathway describes the catabolism of alpha-1,4-glucans of glycogen, starch and maltosaccharides to various metabolites via the central intermediate AF. The reaction sequence of the pathway consists of more than 10 enzymatic steps. This pathway occurs in certain bacteria, fungi, algae and mammals. In this communication, the AF pathway and its regulatory mechanisms in these organisms are presented and the metabolites of this pathway as antioxidants and antimicrobials in biotic and abiotic stress responses and in carbon starvation signaling are discussed.


Subject(s)
Fructose/analogs & derivatives , Fructose/metabolism , Signal Transduction , Animals , Carbohydrate Dehydrogenases/metabolism , Catalysis , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...