Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
2.
Arterioscler Thromb Vasc Biol ; 43(11): 2119-2132, 2023 11.
Article in English | MEDLINE | ID: mdl-37675632

ABSTRACT

BACKGROUND: Regulatory T cells (Tregs) are protective in atherosclerosis but reduced during disease progression due to cell death and loss of stability. However, the mechanisms of Treg dysfunction remain unknown. Oxidized phospholipids are abundant in atherosclerosis and can activate innate immune cells, but little is known regarding their impact on T cells. Given Treg loss during atherosclerosis progression and oxidized phospholipid levels in the plaque microenvironment, we investigated whether oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (oxPAPC), an oxidized phospholipid associated with atherosclerotic plaques, alters Treg differentiation and function. METHODS: CD4+ T cells were polarized to Treg, T helper (Th) 1, and Th17 cells with or without oxPAPC and assessed by flow cytometry. Gene expression in oxPAPC-treated Tregs was analyzed by bulk RNA sequencing. Functional studies of oxPAPC-induced Tregs were performed by coculturing Tregs with CellTrace Violet-labeled cells in vitro, and by adoptively transferring Tregs to hyperlipidemic Ldlr-/- mice to measure atherosclerosis progression. RESULTS: Compared with controls, oxPAPC-treated Tregs were less viable, but surviving cells expressed higher levels of the Th1-associated markers T-bet, CXCR3, and IFN (interferon)-γ. Th1 and Th17 skewing cultures were unaltered by oxPAPC. IFN-γ is linked to Treg instability, thus Treg polarization experiments were repeated using Ifngr1-/- CD4+ T cells. IFNγR1 (INF gamma receptor 1) deficiency did not improve cell viability in oxPAPC-treated Tregs; however, T-bet and IFN-γ expression was not increased in surviving cells suggesting a role for IFN-γsignaling. OxPAPC-treated Tregs were less suppressive in vitro, and adoptive transfer studies in hyperlipidemic Ldlr-/- mice showed that oxPAPC-induced Tregs possessed altered tissue homing and were insufficient to inhibit atherosclerosis progression. CONCLUSIONS: OxPAPC elicits Treg-specific changes altering Treg differentiation and inducing a Th1-like phenotype in surviving cells partially through IFN-γ signaling. This is biologically relevant as oxPAPC-treated Tregs do not reduce atherosclerosis progression in Ldlr-/- mice. This study supports the role of oxidized phospholipids in negatively impacting Treg differentiation and atheroprotective function.


Subject(s)
Atherosclerosis , Phospholipids , Mice , Animals , Phospholipids/metabolism , T-Lymphocytes, Regulatory , Interferon-gamma/metabolism , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Cell Differentiation
3.
Sci Immunol ; 8(79): eabq0178, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36638190

ABSTRACT

T cells in systemic lupus erythematosus (SLE) exhibit multiple metabolic abnormalities. Excess iron can impair mitochondria and may contribute to SLE. To gain insights into this potential role of iron in SLE, we performed a CRISPR screen of iron handling genes on T cells. Transferrin receptor (CD71) was identified as differentially critical for TH1 and inhibitory for induced regulatory T cells (iTregs). Activated T cells induced CD71 and iron uptake, which was exaggerated in SLE-prone T cells. Cell surface CD71 was enhanced in SLE-prone T cells by increased endosomal recycling. Blocking CD71 reduced intracellular iron and mTORC1 signaling, which inhibited TH1 and TH17 cells yet enhanced iTregs. In vivo treatment reduced kidney pathology and increased CD4 T cell production of IL-10 in SLE-prone mice. Disease severity correlated with CD71 expression on TH17 cells from patients with SLE, and blocking CD71 in vitro enhanced IL-10 secretion. T cell iron uptake via CD71 thus contributes to T cell dysfunction and can be targeted to limit SLE-associated pathology.


Subject(s)
Lupus Erythematosus, Systemic , Receptors, Transferrin , T-Lymphocytes, Regulatory , Animals , Mice , Interleukin-10/metabolism , Lupus Erythematosus, Systemic/metabolism , Receptors, Transferrin/metabolism , T-Lymphocytes, Regulatory/metabolism , Humans
4.
Science ; 373(6556): 813-818, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34385401

ABSTRACT

A Western-style, high-fat diet promotes cardiovascular disease, in part because it is rich in choline, which is converted to trimethylamine (TMA) by the gut microbiota. However, whether diet-induced changes in intestinal physiology can alter the metabolic capacity of the microbiota remains unknown. Using a mouse model of diet-induced obesity, we show that chronic exposure to a high-fat diet escalates Escherichia coli choline catabolism by altering intestinal epithelial physiology. A high-fat diet impaired the bioenergetics of mitochondria in the colonic epithelium to increase the luminal bioavailability of oxygen and nitrate, thereby intensifying respiration-dependent choline catabolism of E. coli In turn, E. coli choline catabolism increased levels of circulating trimethlamine N-oxide, which is a potentially harmful metabolite generated by gut microbiota.


Subject(s)
Colon/physiology , Diet, High-Fat , Escherichia coli/metabolism , Intestinal Mucosa/physiology , Methylamines/metabolism , Animals , Cell Hypoxia , Choline/administration & dosage , Choline/metabolism , Colon/cytology , Energy Metabolism , Epithelial Cells/physiology , Escherichia coli/genetics , Escherichia coli/growth & development , Feces/microbiology , Gastrointestinal Microbiome , Inflammation , Intestinal Mucosa/metabolism , Male , Methylamines/blood , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Nitrates/metabolism , Obesity , Oxygen Consumption
5.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34403367

ABSTRACT

Autoimmune disease has presented an insurmountable barrier to restoration of durable immune tolerance. Previous studies indicate that chronic therapy with metabolic inhibitors can reduce autoimmune inflammation, but it remains unknown whether acute metabolic modulation enables permanent immune tolerance to be established. In an animal model of lupus, we determined that targeting glucose metabolism with 2-deoxyglucose (2DG) and mitochondrial metabolism with metformin enables endogenous immune tolerance mechanisms to respond to tolerance induction. A 2-week course of 2DG and metformin, when combined with tolerance-inducing therapy anti-CD45RB, prevented renal deposition of autoantibodies for 6 months after initial treatment and restored tolerance induction to allografts in lupus-prone mice. The restoration of durable immune tolerance was linked to changes in T cell surface glycosylation patterns, illustrating a role for glycoregulation in immune tolerance. These findings indicate that metabolic therapy may be applied as a powerful preconditioning to reinvigorate tolerance mechanisms in autoimmune and transplant settings that resist current immune therapies.


Subject(s)
Antimetabolites/pharmacology , CD4-Positive T-Lymphocytes/immunology , Deoxyglucose/pharmacology , Hypoglycemic Agents/pharmacology , Immune Tolerance/drug effects , Kidney/drug effects , Leukocyte Common Antigens/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Metformin/pharmacology , Animals , Antibodies/pharmacology , Disease Models, Animal , Glycolysis/drug effects , Glycosylation/drug effects , Immune Tolerance/immunology , Kidney/immunology , Leukocyte Common Antigens/antagonists & inhibitors , Mice , Oxidative Phosphorylation/drug effects , Transplantation Tolerance/drug effects , Transplantation Tolerance/immunology , Transplantation, Homologous
6.
Curr Opin Rheumatol ; 33(2): 211-218, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33394753

ABSTRACT

PURPOSE OF REVIEW: Accelerated atherosclerosis is a significant comorbidity and the leading cause of death for patients with systemic lupus erythematosus (SLE). It is now apparent that SLE-accelerated atherosclerosis is not driven solely by traditional cardiovascular risk factors, adding complexity to disease characterization and mechanistic understanding. In this review, we will summarize new insights into SLE-accelerated atherosclerosis evaluation, treatment, and mechanism. RECENT FINDINGS: Recent work highlights the need to incorporate inflammatory biomarkers into cardiovascular disease (CVD) risk assessments. This is especially true for SLE patients, in which mechanisms of immune dysfunction likely drive CVD progression. There is new evidence that commonly prescribed SLE therapeutics hinder atherosclerosis development. This effect is achieved both by reducing SLE-associated inflammation and by directly improving measures of atherosclerosis, emphasizing the interconnected mechanisms of the two conditions. SUMMARY: SLE-accelerated atherosclerosis is most likely the consequence of chronic autoimmune inflammation. Therefore, diligent management of atherosclerosis requires assessment of SLE disease activity as well as traditional cardiovascular risk factors. This supports why many of the therapeutics classically used to control SLE also modulate atherosclerosis development. Greater understanding of the mechanisms underlying this condition will allow for the development of more targeted therapeutics and improved outcomes for SLE patients.


Subject(s)
Atherosclerosis , Lupus Erythematosus, Systemic , Atherosclerosis/epidemiology , Atherosclerosis/etiology , Humans , Inflammation , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/drug therapy , Risk Factors
7.
Int J Nanomedicine ; 15: 1215-1228, 2020.
Article in English | MEDLINE | ID: mdl-32110018

ABSTRACT

BACKGROUND: Helper T cell activity is dysregulated in a number of diseases including those associated with rheumatic autoimmunity. Treatment options are limited and usually consist of systemic immune suppression, resulting in undesirable consequences from compromised immunity. Hedgehog (Hh) signaling has been implicated in the activation of T cells and the formation of the immune synapse, but remains understudied in the context of autoimmunity. Modulation of Hh signaling has the potential to enable controlled immunosuppression but a potential therapy has not yet been developed to leverage this opportunity. METHODS: In this work, we developed biodegradable nanoparticles to enable targeted delivery of eggmanone (Egm), a specific Hh inhibitor, to CD4+ T cell subsets. We utilized two FDA-approved polymers, poly(lactic-co-glycolic acid) and polyethylene glycol, to generate hydrolytically degradable nanoparticles. Furthermore, we employed maleimide-thiol mediated conjugation chemistry to decorate nanoparticles with anti-CD4 F(ab') antibody fragments to enable targeted delivery of Egm. RESULTS: Our novel delivery system achieved a highly specific association with the majority of CD4+ T cells present among a complex cell population. Additionally, we have demonstrated antigen-specific inhibition of CD4+ T cell responses mediated by nanoparticle-formulated Egm. CONCLUSION: This work is the first characterization of Egm's immunomodulatory potential. Importantly, this study also suggests the potential benefit of a biodegradable delivery vehicle that is rationally designed for preferential interaction with a specific immune cell subtype for targeted modulation of Hh signaling.


Subject(s)
Autoimmunity/drug effects , Drug Delivery Systems/methods , Immunologic Factors/administration & dosage , Nanoparticles/administration & dosage , Pyrimidinones/administration & dosage , T-Lymphocytes/drug effects , Thiophenes/administration & dosage , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Immunoglobulin Fragments/chemistry , Mice, Inbred C57BL , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rheumatic Diseases/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology
8.
Atherosclerosis ; 285: 108-119, 2019 06.
Article in English | MEDLINE | ID: mdl-31051414

ABSTRACT

BACKGROUND AND AIMS: Circulating levels of oxidized lipoprotein (oxLDL) correlate with myocardial infarction risk and atherosclerosis severity. Our previous study demonstrates that oxLDL immune complexes (oxLDL-ICs) can signal through FcγRs on bone marrow-derived dendritic cells (BMDCs) and enhance their activation and inflammatory cytokine secretion. While global FcγR-/- studies have shown that activating FcγRs are proatherogenic, the role of the inhibitory FcγRIIb is unclear. We sought to determine the role of DC-specific FcγRIIb in atherosclerosis. METHODS: Bone marrow chimeras were generated by rescuing lethally irradiated Ldlr-/- mice with hematopoietic cells from littermate CD11c-Cre+ or CD11c-Cre-Fcgr2bfl/fl donors. Four weeks following transplant, recipients were placed on a Western diet for eight weeks. Various tissues and organs were analyzed for differences in inflammation. RESULTS: Quantitation of atherosclerosis in the proximal aorta demonstrated a 58% increase in female CD11c-Cre+Fcgr2bfl/fl recipients, but a surprising 44% decrease in male recipients. Hepatic cholesterol and triglycerides were increased in female CD11c-Cre+Fcgr2bfl/fl recipients. This was associated with an increase in CD36 and MHC Class II expression on hepatic CD11c+CD11b+ DCs in female livers. In contrast, male CD11c-Cre+Fcgr2bfl/fl recipients had decreased hepatic lipids with a corresponding decrease in CD36 and MHC Class II expression on CD11c+ cells. Interestingly, both sexes of CD11c-Cre+Fcgr2bfl/fl recipients had significant decreases in serum cholesterol and TGs with corresponding decreases in liver Fasn transcripts. CONCLUSIONS: The absence of FcγRIIb expression on CD11c+ cells results in sex-dependent alteration in liver inflammation influencing atherogenesis and sex-independent modulation of serum cholesterol and TGs.


Subject(s)
Atherosclerosis/blood , Cholesterol/blood , Receptors, IgG/physiology , Triglycerides/blood , Animals , CD11 Antigens/biosynthesis , Dendritic Cells/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Receptors, IgG/biosynthesis , Sex Factors
9.
Crit Rev Immunol ; 38(4): 333-342, 2018.
Article in English | MEDLINE | ID: mdl-30806246

ABSTRACT

Cardiovascular disease (CVD) is the number one cause of death in the United States and worldwide. The most common cause of cardiovascular disease is atherosclerosis, or formation of fatty plaques in the arteries. Low-density lipoprotein (LDL), termed "bad cholesterol", is a large molecule comprised of many proteins as well as lipids including cholesterol, phospholipids, and triglycerides. Circulating levels of LDL are directly associated with atherosclerosis disease severity. Once thought to simply be caused by passive retention of LDL in the vasculature, atherosclerosis studies over the past 40-50 years have uncovered a much more complex mechanism. It has now become well established that within the vasculature, LDL can undergo many different types of oxidative modifications such as esterification and lipid peroxidation. The resulting oxidized LDL (oxLDL) has been found to have antigenic potential and contribute heavily to atherosclerosis associated inflammation, activating both innate and adaptive immunity. This review discusses the many proposed mechanisms by which oxidized LDL modulates inflammatory responses and how this might modulate atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Inflammation/immunology , Inflammation/pathology , Lipoproteins, LDL/immunology , Animals , Atherosclerosis/blood , Atherosclerosis/immunology , Humans , Inflammation/blood , Lipoproteins, LDL/blood
10.
Nat Rev Rheumatol ; 13(5): 313-320, 2017 May.
Article in English | MEDLINE | ID: mdl-28381829

ABSTRACT

All immune cells depend on specific and efficient metabolic pathways to mount an appropriate response. Over the past decade, the field of immunometabolism has expanded our understanding of the various means by which cells modulate metabolism to achieve the effector functions necessary to fight infection or maintain homeostasis. Harnessing these metabolic pathways to manipulate inappropriate immune responses as a therapeutic strategy in cancer and autoimmunity has received increasing scrutiny by the scientific community. Fine tuning immunometabolism to provide the desired response, or prevent a deleterious response, is an attractive alternative to chemotherapy or overt immunosuppression. The various metabolic pathways used by immune cells in rheumatoid arthritis, systemic lupus erythematosus and osteoarthritis offer numerous opportunities for selective targeting of specific immune cell subsets to manipulate cellular metabolism for therapeutic benefit in these rheumatologic diseases.


Subject(s)
Rheumatic Diseases/immunology , Rheumatic Diseases/therapy , Animals , Drug Therapy , Energy Metabolism , Homeostasis , Humans , Immunotherapy , Metabolic Networks and Pathways , Rheumatic Diseases/metabolism
11.
J Immunol ; 198(5): 2105-2114, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28130494

ABSTRACT

Oxidized low-density lipoprotein (oxLDL) is known to activate inflammatory responses in a variety of cells, especially macrophages and dendritic cells. Interestingly, much of the oxLDL in circulation is complexed to Abs, and these resulting immune complexes (ICs) are a prominent feature of chronic inflammatory disease, such as atherosclerosis, type-2 diabetes, systemic lupus erythematosus, and rheumatoid arthritis. Levels of oxLDL ICs often correlate with disease severity, and studies demonstrated that oxLDL ICs elicit potent inflammatory responses in macrophages. In this article, we show that bone marrow-derived dendritic cells (BMDCs) incubated with oxLDL ICs for 24 h secrete significantly more IL-1ß compared with BMDCs treated with free oxLDL, whereas there was no difference in levels of TNF-α or IL-6. Treatment of BMDCs with oxLDL ICs increased expression of inflammasome-related genes Il1a, Il1b, and Nlrp3, and pretreatment with a caspase 1 inhibitor decreased IL-1ß secretion in response to oxLDL ICs. This inflammasome priming was due to oxLDL IC signaling via multiple receptors, because inhibition of CD36, TLR4, and FcγR significantly decreased IL-1ß secretion in response to oxLDL ICs. Signaling through these receptors converged on the adaptor protein CARD9, a component of the CARD9-Bcl10-MALT1 signalosome complex involved in NF-κB translocation. Finally, oxLDL IC-mediated IL-1ß production resulted in increased Th17 polarization and cytokine secretion. Collectively, these data demonstrate that oxLDL ICs induce inflammasome activation through a separate and more robust mechanism than oxLDL alone and that these ICs may be immunomodulatory in chronic disease and not just biomarkers of severity.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Dendritic Cells/immunology , Inflammasomes/metabolism , Lipoproteins, LDL/metabolism , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Th17 Cells/immunology , Animals , Antibodies/metabolism , Antigen-Antibody Complex/metabolism , Cells, Cultured , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptor Cross-Talk , Receptors, IgG/metabolism , Signal Transduction , Toll-Like Receptors/metabolism
12.
Arterioscler Thromb Vasc Biol ; 36(8): 1483-95, 2016 08.
Article in English | MEDLINE | ID: mdl-27365402

ABSTRACT

OBJECTIVE: Antiatherosclerotic effects of tumor necrosis factor-α (TNF-α) blockade in patients with systemic inflammatory states are not conclusively demonstrated, which suggests that effects depend on the cause of inflammation. Macrophage LRP1 (low-density lipoprotein receptor-related protein 1) and apoE contribute to inflammation through different pathways. We studied the antiatherosclerosis effects of TNF-α blockade in hyperlipidemic mice lacking either LRP1 (MΦLRP1(-/-)) or apoE from macrophages. APPROACH AND RESULTS: Lethally irradiated low-density lipoprotein receptor (LDLR)(-/-) mice were reconstituted with bone marrow from either wild-type, MΦLRP1(-/-), apoE(-/-) or apoE(-/-)/MΦLRP1(-/-)(DKO) mice, and then treated with the TNF-α inhibitor adalimumab while fed a Western-type diet. Adalimumab reduced plasma TNF-α concentration, suppressed blood ly6C(hi) monocyte levels and their migration into the lesion, and reduced lesion cellularity and inflammation in both wild-type→LDLR(-/-) and apoE(-/-)→LDLR(-/-) mice. Overall, adalimumab reduced lesion burden by 52% to 57% in these mice. Adalimumab reduced TNF-α and blood ly6C(hi) monocyte levels in MΦLRP1(-/-)→LDLR(-/-) and DKO→LDLR(-/-) mice, but it did not suppress ly6C(hi) monocyte migration into the lesion or atherosclerosis progression. CONCLUSIONS: Our results show that TNF-α blockade exerts antiatherosclerotic effects that are dependent on the presence of macrophage LRP1.


Subject(s)
Adalimumab/pharmacology , Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Drug Resistance , Macrophages/drug effects , Receptors, LDL/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Animals , Antigens, Ly/metabolism , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Transplantation , Cell Movement/drug effects , Diet, High-Fat , Disease Models, Animal , Drug Resistance/genetics , Female , Genetic Predisposition to Disease , Low Density Lipoprotein Receptor-Related Protein-1 , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Necrosis , Phenotype , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Whole-Body Irradiation
13.
J Pathol ; 238(1): 52-62, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26333678

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes atherosclerosis by increasing low-density lipoprotein (LDL) cholesterol levels through degradation of hepatic LDL receptor (LDLR). Studies have described the systemic effects of PCSK9 on atherosclerosis, but whether PCSK9 has local and direct effects on the plaque is unknown. To study the local effect of human PCSK9 (hPCSK9) on atherosclerotic lesion composition, independently of changes in serum cholesterol levels, we generated chimeric mice expressing hPCSK9 exclusively from macrophages, using marrow from hPCSK9 transgenic (hPCSK9tg) mice transplanted into apoE(-/-) and LDLR(-/-) mice, which were then placed on a high-fat diet (HFD) for 8 weeks. We further characterized the effect of hPCSK9 expression on the inflammatory responses in the spleen and by mouse peritoneal macrophages (MPM) in vitro. We found that MPMs from transgenic mice express both murine (m) Pcsk9 and hPCSK9 and that the latter reduces macrophage LDLR and LRP1 surface levels. We detected hPCSK9 in the serum of mice transplanted with hPCSK9tg marrow, but did not influence lipid levels or atherosclerotic lesion size. However, marrow-derived PCSK9 progressively accumulated in lesions of apoE(-/-) recipient mice, while increasing the infiltration of Ly6C(hi) inflammatory monocytes by 32% compared with controls. Expression of hPCSK9 also increased CD11b- and Ly6C(hi) -positive cell numbers in spleens of apoE(-/-) mice. In vitro, expression of hPCSK9 in LPS-stimulated macrophages increased mRNA levels of the pro-inflammatory markers Tnf and Il1b (40% and 45%, respectively) and suppressed those of the anti-inflammatory markers Il10 and Arg1 (30% and 44%, respectively). All PCSK9 effects were LDLR-dependent, as PCSK9 protein was not detected in lesions of LDLR(-/-) recipient mice and did not affect macrophage or splenocyte inflammation. In conclusion, PCSK9 directly increases atherosclerotic lesion inflammation in an LDLR-dependent but cholesterol-independent mechanism, suggesting that therapeutic PCSK9 inhibition may have vascular benefits secondary to LDL reduction.


Subject(s)
Atherosclerosis/metabolism , Macrophages, Peritoneal/metabolism , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Animals , Atherosclerosis/pathology , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proprotein Convertase 9 , Receptors, LDL/metabolism , Transplantation Chimera
14.
Ann Rheum Dis ; 74(4): 778-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24395554

ABSTRACT

OBJECTIVE: Accelerated atherosclerosis is a major source of morbidity in systemic lupus erythematosus (SLE). However, the cause of SLE-accelerated atherosclerosis remains unclear. METHODS: CD4(+) T cells from C57/Bl/6 (B6) or SLE-susceptible B6.Sle1.2.3 (B6.SLE) mice were transferred into LDLr(-/-), Rag(-/-) mice. T cells were examined for cytokine production and expression of interleukin-10 receptor (IL-10R) and functional markers. T cells were isolated based on FoxP3(GFP) expression and transferred to LDLr(-/-), Rag(-/-) mice to establish a role for B6.SLE effector T cells (Teff) in atherosclerosis. RESULTS: Mice receiving whole B6.SLE CD4(+) T cells displayed no other SLE phenotype; however, atherosclerosis was increased nearly 40%. We noted dysregulated IL-17 production and reduced frequency of IL-10R expression by B6.SLE regulatory T cells (Treg). Functional assays indicated resistance of B6.SLE Teff to suppression by both B6.SLE and B6 Treg. Transfer experiments with CD4(+)FoxP3(-) Teff and CD4(+)FoxP3(+) Treg from B6.SLE and B6 mice, respectively, resulted in increased atherosclerosis compared with B6 Teff and Treg recipients. Treg isolated from mice receiving B6.SLE Teff with B6 Treg had increased production of IL-17 and fewer expressed IL-10R compared with B6 Teff and Treg transfer. CONCLUSIONS: Transfer of B6.SLE Teff to LDLr(-/-), Rag(-/-) mice results in accelerated atherosclerosis independent of the source of Treg. In addition, the presence of B6.SLE Teff resulted in more IL-17-producing Treg and fewer expressing IL-10R, suggesting that B6.SLE Teff may mediate phenotypic changes in Treg. To our knowledge, this is the first study to provide direct evidence of the role of B6.SLE Teff in accelerating atherosclerosis through resistance to Treg suppression.


Subject(s)
Atherosclerosis/metabolism , Lupus Erythematosus, Systemic/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred Strains , Mice, Knockout , Receptors, Interleukin-10/metabolism , Receptors, LDL/genetics , T-Lymphocyte Subsets , T-Lymphocytes, Regulatory/immunology
15.
PLoS One ; 9(7): e102236, 2014.
Article in English | MEDLINE | ID: mdl-25050824

ABSTRACT

Expression of molecules involved in lipid homeostasis such as the low density lipoprotein receptor (LDLr) on antigen presenting cells (APCs) has been shown to enhance invariant natural killer T (iNKT) cell function. However, the contribution to iNKT cell activation by other lipoprotein receptors with shared structural and ligand binding properties to the LDLr has not been described. In this study, we investigated whether a structurally related receptor to the LDLr, known as LDL receptor-related protein (LRP), plays a role in iNKT cell activation. We found that, unlike the LDLr which is highly expressed on all immune cells, the LRP was preferentially expressed at high levels on F4/80+ macrophages (MΦ). We also show that CD169+ MΦs, known to present antigen to iNKT cells, exhibited increased expression of LRP compared to CD169- MΦs. To test the contribution of MΦ LRP to iNKT cell activation we used a mouse model of MΦ LRP conditional knockout (LRP-cKO). LRP-cKO MΦs pulsed with glycolipid alpha-galactosylceramide (αGC) elicited normal IL-2 secretion by iNKT hybridoma and in vivo challenge of LRP-cKO mice led to normal IFN-γ, but blunted IL-4 response in both serum and intracellular expression by iNKT cells. Flow cytometric analyses show similar levels of MHC class-I like molecule CD1d on LRP-cKO MΦs and normal glycolipid uptake. Survey of the iNKT cell compartment in LRP-cKO mice revealed intact numbers and percentages and no homeostatic disruption as evidenced by the absence of programmed death-1 and Ly-49 surface receptors. Mixed bone marrow chimeras showed that the inability iNKT cells to make IL-4 is cell extrinsic and can be rescued in the presence of wild type APCs. Collectively, these data demonstrate that, although MΦ LRP may not be necessary for IFN-γ responses, it can contribute to iNKT cell activation by enhancing early IL-4 secretion.


Subject(s)
Cytokines/biosynthesis , Macrophages/metabolism , Natural Killer T-Cells/metabolism , Receptors, LDL/genetics , Tumor Suppressor Proteins/genetics , Adaptive Immunity , Animals , Antigens, CD1d/metabolism , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Gene Knockout Techniques , Immunoglobulin E/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism
16.
Adipocyte ; 3(2): 141-5, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24719788

ABSTRACT

The field of immunometabolism is burgeoning, with hundreds of papers published on the topic each year. Our understanding of the contribution of immune cells to metabolic regulation has expanded from a simple idea of innate immune cells, such as macrophages, altering adipose tissue function in obesity, to an awareness of the complex role of adaptive immunity in many different organ systems. Recent findings have clearly demonstrated the presence of adaptive lymphocytes, such as T and B cells, in adipose tissue. Furthermore, these data demonstrated T-cell accumulation and limited T-cell receptor repertoire diversity in obese adipose tissue, indicating that an antigen-specific immune response may occur within this tissue. In a recently published paper, we reported that a mouse model of weight cycling resulted in increased T-cell accumulation in adipose tissue. In the current commentary, we discuss the possibility that this increase in adipose tissue T-cell number could represent a local secondary immune response to self-antigens exposed in adipose tissue during obesity. If further experimentation indicates that this hypothesis is true, these data will fortify the concept that obesity is a complex immune-mediated disease and would emphasize the importance of designing therapies to maintain weight loss.

17.
PLoS One ; 8(11): e80089, 2013.
Article in English | MEDLINE | ID: mdl-24223214

ABSTRACT

OBJECTIVE: MCPIP1 is a newly identified protein that profoundly impacts immunity and inflammation. We aim to test if MCPIP1 deficiency in hematopoietic cells results in systemic inflammation and accelerates atherogenesis in mice. APPROACH AND RESULTS: After lethally irradiated, LDLR(-/-) mice were transplanted with bone marrow cells from either wild-type or MCPIP1(-/-) mice. These chimeric mice were fed a western-type diet for 7 weeks. We found that bone marrow MCPIP1(-/-) mice displayed a phenotype similar to that of whole body MCPIP1(-/-) mice, with severe systemic and multi-organ inflammation. However, MCPIP1(-/-) bone marrow recipients developed >10-fold less atherosclerotic lesions in the proximal aorta than WT bone marrow recipients, and essentially no lesions in en face aorta. The diminishment in atherosclerosis in bone marrow MCPIP1(-/-) mice may be partially attributed to the slight decrease in their plasma lipids. Flow cytometric analysis of splenocytes showed that bone marrow MCPIP1(-/-) mice contained reduced numbers of T cells and B cells, but increased numbers of regulatory T cells, Th17 cells, CD11b+/Gr1+ cells and CD11b+/Ly6C(low) cells. This overall anti-atherogenic leukocyte profile may also contribute to the reduced atherogenesis. We also examined the cholesterol efflux capability of MCPIP1 deficient macrophages, and found that MCPIP1 deficiency increased cholesterol efflux to apoAI and HDL, due to increased protein levels of ABCA1 and ABCG1. CONCLUSIONS: Hematopoietic deficiency of MCPIP1 resulted in severe systemic and multi-organ inflammation but paradoxically diminished atherogenesis in mice. The reduced atheroegensis may be explained by the decreased plasma cholesterol levels, the anti-atherogenic leukocyte profile, as well as enhanced cholesterol efflux capability. This study suggests that, while atherosclerosis is a chronic inflammatory disease, the mechanisms underlying atherogenesis-associated inflammation in arterial wall versus the inflammation in solid organs may be substantially different.


Subject(s)
Atherosclerosis/metabolism , Bone Marrow/metabolism , Hyperlipidemias/metabolism , Inflammation/metabolism , Ribonucleases/deficiency , Animals , Atherosclerosis/genetics , Bone Marrow Transplantation , Female , Hyperlipidemias/genetics , Inflammation/genetics , Mice , Mice, Knockout , Ribonucleases/genetics
19.
J Am Heart Assoc ; 2(2): e000093, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23563994

ABSTRACT

BACKGROUND: Elevated cholesterol and triglycerides in blood lead to atherosclerosis and fatty liver, contributing to rising cardiovascular and hepatobiliary morbidity and mortality worldwide. METHODS AND RESULTS: A cell-penetrating nuclear transport modifier (NTM) reduced hyperlipidemia, atherosclerosis, and fatty liver in low-density lipoprotein receptor-deficient mice fed a Western diet. NTM treatment led to lower cholesterol and triglyceride levels in blood compared with control animals (36% and 53%, respectively; P<0.005) and liver (41% and 34%, respectively; P<0.05) after 8 weeks. Atherosclerosis was reduced by 63% (P<0.0005), and liver function improved compared with saline-treated controls. In addition, fasting blood glucose levels were reduced from 209 to 138 mg/dL (P<0.005), and body weight gain was ameliorated (P<0.005) in NTM-treated mice, although food intake remained the same as that in control animals. The NTM used in this study, cSN50.1 peptide, is known to modulate nuclear transport of stress-responsive transcription factors such as nuclear factor kappa B, the master regulator of inflammation. This NTM has now been demonstrated to also modulate nuclear transport of sterol regulatory element-binding protein (SREBP) transcription factors, the master regulators of cholesterol, triglyceride, and fatty acid synthesis. NTM-modulated translocation of SREBPs to the nucleus was associated with attenuated transactivation of their cognate genes that contribute to hyperlipidemia. CONCLUSIONS: Two-pronged control of inflammation and dyslipidemia by modulating nuclear transport of their critical regulators offers a new approach to comprehensive amelioration of hyperlipidemia, atherosclerosis, fatty liver, and their potential complications.


Subject(s)
Atherosclerosis/drug therapy , Cell Nucleus/metabolism , Cell-Penetrating Peptides/therapeutic use , Fatty Liver/drug therapy , Hypercholesterolemia/drug therapy , NF-kappa B/metabolism , Peptides/therapeutic use , Sterol Regulatory Element Binding Proteins/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Atherosclerosis/metabolism , Cell Nucleus/drug effects , Cell-Penetrating Peptides/pharmacology , Cholesterol/metabolism , Dietary Fats/metabolism , Disease Models, Animal , Fatty Liver/metabolism , Female , Hypercholesterolemia/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Transgenic , NF-kappa B/drug effects , Peptides/pharmacology , Sterol Regulatory Element Binding Proteins/drug effects , Transcription Factors/drug effects , Transcription Factors/metabolism , Triglycerides/metabolism
20.
Am J Physiol Endocrinol Metab ; 303(11): E1313-24, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23032686

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies against self-antigens such as double-stranded DNA and phospholipids. Classical comorbidities of SLE include glomerulonephritis, infection, cardiovascular disease, arthritis, skin disorders, and neurological disease. In addition to these classical comorbidities, there is emerging evidence that SLE patients are at higher risk of developing insulin resistance and other components of the metabolic syndrome. Visceral adipose tissue inflammation is a central mediator of insulin resistance in the obese setting, but the mechanism behind the pathogenesis of metabolic disease in the SLE patient population is unclear. We hypothesize that lupus-associated changes in the adaptive immune system are associated with disruption in glucose homeostasis in the context of SLE. To test this hypothesis, we assessed the metabolic and immunological phenotype of SLE-prone B6.SLE mice. B6.SLE mice fed a low-fat diet had significantly worsened glucose tolerance, increased adipose tissue insulin resistance, increased ß-cell insulin secretion, and increased adipocyte size compared with their respective B6 controls. Independently of diet, B cells isolated from the white adipose tissue of B6.SLE mice were skewed toward IgG production, and the level of IgG1 was elevated in the serum of SLE-prone mice. These data show that B6.SLE mice develop defects in glucose homeostasis even when fed a low-fat diet and suggest that B cells may play a role in this metabolic dysfunction.


Subject(s)
B-Lymphocytes/immunology , Insulin Resistance/physiology , Insulin/blood , Lupus Erythematosus, Systemic/metabolism , Metabolic Syndrome/complications , Adaptive Immunity/physiology , Adipocytes/cytology , Adipose Tissue/immunology , Analysis of Variance , Animals , Disease Models, Animal , Glucose Tolerance Test , Homeostasis/immunology , Homeostasis/physiology , Insulin Resistance/immunology , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/physiopathology , Metabolic Syndrome/blood , Metabolic Syndrome/immunology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...