Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 999021, 2022.
Article in English | MEDLINE | ID: mdl-36189251

ABSTRACT

AAV gene transfer is a promising treatment for many patients with life-threatening genetic diseases. However, host immune response to the vector poses a significant challenge for the durability and safety of AAV-mediated gene therapy. Here, we characterize the innate immune response to AAV in human whole blood. We identified neutrophils, monocyte-related dendritic cells, and monocytes as the most prevalent cell subsets able to internalize AAV particles, while conventional dendritic cells were the most activated in terms of the CD86 co-stimulatory molecule upregulation. Although low titers (≤1:10) of AAV neutralizing antibodies (NAb) in blood did not have profound effects on the innate immune response to AAV, higher NAb titers (≥1:100) significantly increased pro-inflammatory cytokine/chemokine secretion, vector uptake by antigen presenting cells (APCs) and complement activation. Interestingly, both full and empty viral particles were equally potent in inducing complement activation and cytokine secretion. By using a compstatin-based C3 and C3b inhibitor, APL-9, we demonstrated that complement pathway inhibition lowered CD86 levels on APCs, AAV uptake, and cytokine/chemokine secretion in response to AAV. Together these results suggest that the pre-existing humoral immunity to AAV may contribute to trigger adverse immune responses observed in AAV-based gene therapy, and that blockade of complement pathway may warrant further investigation as a potential strategy for decreasing immunogenicity of AAV-based therapeutics.


Subject(s)
Dependovirus , Genetic Vectors , Antibodies, Neutralizing , Cytokines/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Humans , Immunity, Humoral
2.
Nat Commun ; 12(1): 6393, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737297

ABSTRACT

Pompe disease (PD) is a severe neuromuscular disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). PD is currently treated with enzyme replacement therapy (ERT) with intravenous infusions of recombinant human GAA (rhGAA). Although the introduction of ERT represents a breakthrough in the management of PD, the approach suffers from several shortcomings. Here, we developed a mouse model of PD to compare the efficacy of hepatic gene transfer with adeno-associated virus (AAV) vectors expressing secretable GAA with long-term ERT. Liver expression of GAA results in enhanced pharmacokinetics and uptake of the enzyme in peripheral tissues compared to ERT. Combination of gene transfer with pharmacological chaperones boosts GAA bioavailability, resulting in improved rescue of the PD phenotype. Scale-up of hepatic gene transfer to non-human primates also successfully results in enzyme secretion in blood and uptake in key target tissues, supporting the ongoing clinical translation of the approach.


Subject(s)
Glycogen Storage Disease Type II/enzymology , alpha-Glucosidases/metabolism , Animals , Autophagy , Enzyme Replacement Therapy , Female , Glycogen Storage Disease Type II/therapy , Liver/enzymology , Male , Mice , alpha-Glucosidases/genetics
3.
Cell Metab ; 33(5): 905-922.e6, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33887198

ABSTRACT

Low-protein diets promote metabolic health in rodents and humans, and the benefits of low-protein diets are recapitulated by specifically reducing dietary levels of the three branched-chain amino acids (BCAAs), leucine, isoleucine, and valine. Here, we demonstrate that each BCAA has distinct metabolic effects. A low isoleucine diet reprograms liver and adipose metabolism, increasing hepatic insulin sensitivity and ketogenesis and increasing energy expenditure, activating the FGF21-UCP1 axis. Reducing valine induces similar but more modest metabolic effects, whereas these effects are absent with low leucine. Reducing isoleucine or valine rapidly restores metabolic health to diet-induced obese mice. Finally, we demonstrate that variation in dietary isoleucine levels helps explain body mass index differences in humans. Our results reveal isoleucine as a key regulator of metabolic health and the adverse metabolic response to dietary BCAAs and suggest reducing dietary isoleucine as a new approach to treating and preventing obesity and diabetes.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Diet , Isoleucine/metabolism , Valine/metabolism , Adipose Tissue, White/metabolism , Animals , Body Mass Index , Diet/veterinary , Energy Metabolism , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Liver/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Protein Serine-Threonine Kinases/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
4.
Cell Rep ; 33(11): 108500, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326785

ABSTRACT

Immune cell function is influenced by metabolic conditions. Low-glucose, high-lactate environments, such as the placenta, gastrointestinal tract, and the tumor microenvironment, are immunosuppressive, especially for glycolysis-dependent effector T cells. We report that nicotinamide adenine dinucleotide (NAD+), which is reduced to NADH by lactate dehydrogenase in lactate-rich conditions, is a key point of metabolic control in T cells. Reduced NADH is not available for NAD+-dependent enzymatic reactions involving glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and 3-phosphoglycerate dehydrogenase (PGDH). We show that increased lactate leads to a block at GAPDH and PGDH, leading to the depletion of post-GAPDH glycolytic intermediates, as well as the 3-phosphoglycerate derivative serine that is known to be important for T cell proliferation. Supplementing serine rescues the ability of T cells to proliferate in the presence of lactate-induced reductive stress. Directly targeting the redox state may be a useful approach for developing novel immunotherapies in cancer and therapeutic immunosuppression.


Subject(s)
Lactic Acid/metabolism , NAD/metabolism , Cell Proliferation , Humans , Oxidation-Reduction
5.
Elife ; 92020 11 10.
Article in English | MEDLINE | ID: mdl-33170774

ABSTRACT

The Pro47Ser variant of p53 (S47) exists in African-descent populations and is associated with increased cancer risk in humans and mice. Due to impaired repression of the cystine importer Slc7a11, S47 cells show increased glutathione (GSH) accumulation compared to cells with wild -type p53. We show that mice containing the S47 variant display increased mTOR activity and oxidative metabolism, as well as larger size, improved metabolic efficiency, and signs of superior fitness. Mechanistically, we show that mTOR and its positive regulator Rheb display increased association in S47 cells; this is due to an altered redox state of GAPDH in S47 cells that inhibits its ability to bind and sequester Rheb. Compounds that decrease glutathione normalize GAPDH-Rheb complexes and mTOR activity in S47 cells. This study reveals a novel layer of regulation of mTOR by p53, and raises the possibility that this variant may have been selected for in early Africa.


Subject(s)
TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Amino Acid Substitution/genetics , Animals , Black People/genetics , Cell Line , Glutathione/metabolism , Glycolysis , Humans , Mitochondria/metabolism , Oxidation-Reduction , TOR Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/metabolism
6.
Front Physiol ; 10: 1439, 2019.
Article in English | MEDLINE | ID: mdl-31849697

ABSTRACT

Lack of interleukin 15 receptor alpha (IL15RA) increases spontaneous activity, exercise capacity and protects from diet-induced obesity by enhancing muscle energy metabolism, suggesting a role as exercise mimetic for IL15RA antagonists. Using controlled in vivo muscle stimulation mimicking moderate exercise in normal and Il15ra-/- mice, we mapped and contrasted the metabolic pathways activated upon stimulation or deletion of IL15RA. Stimulation caused the differential regulation of 123 out of the 321 detected metabolites (FDR ≤ 0.05 and fold change ≥ ±1.5). The main energy pathways activated were fatty acid oxidation, nucleotide metabolism, and anaplerotic reactions. Notably, resting Il15ra-/- muscles were primed in a semi-exercised state, characterized by higher pool sizes of fatty acids oxidized to support muscle activity. These studies identify the role of IL15RA in the system-wide metabolic response to exercise and should enable translational studies to harness the potential of IL15RA blockade as a novel exercise mimetic strategy.

7.
Cell Rep ; 26(13): 3709-3725.e7, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917323

ABSTRACT

Mitochondrial Ca2+ uniporter (MCU)-mediated Ca2+ uptake promotes the buildup of reducing equivalents that fuel oxidative phosphorylation for cellular metabolism. Although MCU modulates mitochondrial bioenergetics, its function in energy homeostasis in vivo remains elusive. Here we demonstrate that deletion of the Mcu gene in mouse liver (MCUΔhep) and in Danio rerio by CRISPR/Cas9 inhibits mitochondrial Ca2+ (mCa2+) uptake, delays cytosolic Ca2+ (cCa2+) clearance, reduces oxidative phosphorylation, and leads to increased lipid accumulation. Elevated hepatic lipids in MCUΔhep were a direct result of extramitochondrial Ca2+-dependent protein phosphatase-4 (PP4) activity, which dephosphorylates AMPK. Loss of AMPK recapitulates hepatic lipid accumulation without changes in MCU-mediated Ca2+ uptake. Furthermore, reconstitution of active AMPK, or PP4 knockdown, enhances lipid clearance in MCUΔhep hepatocytes. Conversely, gain-of-function MCU promotes rapid mCa2+ uptake, decreases PP4 levels, and reduces hepatic lipid accumulation. Thus, our work uncovers an MCU/PP4/AMPK molecular cascade that links Ca2+ dynamics to hepatic lipid metabolism.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Hepatocytes/metabolism , Lipid Metabolism , Mitochondrial Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Calcium Channels/genetics , Cells, Cultured , Female , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Proteins/genetics , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , Zebrafish
8.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135317

ABSTRACT

Alterations in the synthesis and bioavailability of NO are central to the pathogenesis of cardiovascular and metabolic disorders. Although endothelial NO synthase-derived (eNOS-derived) NO affects mitochondrial long-chain fatty acid ß-oxidation, the pathophysiological significance of this regulation remains unclear. Accordingly, we determined the contributions of eNOS/NO signaling in the adaptive metabolic responses to fasting and in age-induced metabolic dysfunction. Four-month-old eNOS-/- mice are glucose intolerant and exhibit serum dyslipidemia and decreased capacity to oxidize fatty acids. However, during fasting, eNOS-/- mice redirect acetyl-CoA to ketogenesis to elevate circulating levels of ß-hydroxybutyrate similar to wild-type mice. Treatment of 4-month-old eNOS-/- mice with nitrite for 10 days corrected the hypertension and serum hyperlipidemia and normalized the rate of fatty acid oxidation. Fourteen-month-old eNOS-/- mice exhibited metabolic derangements, resulting in reduced utilization of fat to generate energy, lower resting metabolic activity, and diminished physical activity. Seven-month administration of nitrite to eNOS-/- mice reversed the age-dependent metabolic derangements and restored physical activity. While the eNOS/NO signaling is not essential for the metabolic adaptation to fasting, it is critical for regulating systemic metabolic homeostasis in aging. The development of age-dependent metabolic disorder is prevented by low-dose replenishment of bioactive NO.


Subject(s)
Aging/metabolism , Homeostasis/drug effects , Nitric Oxide Synthase Type III/deficiency , Sodium Nitrite/administration & dosage , Administration, Oral , Aging/drug effects , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Fasting/metabolism , Humans , Hyperlipidemias/drug therapy , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Hypertension/drug therapy , Hypertension/genetics , Hypertension/metabolism , Male , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Signal Transduction/drug effects , Time Factors , Treatment Outcome
9.
Cell Metab ; 27(5): 1067-1080.e5, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29685734

ABSTRACT

The redox cofactor nicotinamide adenine dinucleotide (NAD) plays a central role in metabolism and is a substrate for signaling enzymes including poly-ADP-ribose-polymerases (PARPs) and sirtuins. NAD concentration falls during aging, which has triggered intense interest in strategies to boost NAD levels. A limitation in understanding NAD metabolism has been reliance on concentration measurements. Here, we present isotope-tracer methods for NAD flux quantitation. In cell lines, NAD was made from nicotinamide and consumed largely by PARPs and sirtuins. In vivo, NAD was made from tryptophan selectively in the liver, which then excreted nicotinamide. NAD fluxes varied widely across tissues, with high flux in the small intestine and spleen and low flux in the skeletal muscle. Intravenous administration of nicotinamide riboside or mononucleotide delivered intact molecules to multiple tissues, but the same agents given orally were metabolized to nicotinamide in the liver. Thus, flux analysis can reveal tissue-specific NAD metabolism.


Subject(s)
Liver/metabolism , NAD/analysis , NAD/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Sirtuins/metabolism , Tryptophan/metabolism , Animals , Female , HCT116 Cells , Hep G2 Cells , Humans , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , NAD/biosynthesis , Niacinamide/administration & dosage , Niacinamide/pharmacokinetics , Spleen/metabolism
10.
J Clin Invest ; 127(11): 4207-4215, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29035283

ABSTRACT

Liver triacylglycerol (TAG) synthesis and secretion are closely linked to nutrient availability. After a meal, hepatic TAG formation from fatty acids is decreased, largely due to a reduction in circulating free fatty acids (FFA). Despite the postprandial decrease in FFA-driven esterification and oxidation, VLDL-TAG secretion is maintained to support peripheral lipid delivery and metabolism. The regulatory mechanisms underlying the postprandial control of VLDL-TAG secretion remain unclear. Here, we demonstrated that the mTOR complex 1 (mTORC1) is essential for this sustained VLDL-TAG secretion and lipid homeostasis. In murine models, the absence of hepatic mTORC1 reduced circulating TAG, despite hepatosteatosis, while activation of mTORC1 depleted liver TAG stores. Additionally, mTORC1 promoted TAG secretion by regulating phosphocholine cytidylyltransferase α (CCTα), the rate-limiting enzyme involved in the synthesis of phosphatidylcholine (PC). Increasing PC synthesis in mice lacking mTORC1 rescued hepatosteatosis and restored TAG secretion. These data identify mTORC1 as a major regulator of phospholipid biosynthesis and subsequent VLDL-TAG secretion, leading to increased postprandial TAG secretion.


Subject(s)
Lipogenesis , Mechanistic Target of Rapamycin Complex 1/physiology , Phosphatidylcholines/biosynthesis , Triglycerides/metabolism , Animals , Cells, Cultured , Fatty Liver/metabolism , Hepatocytes/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Phosphatidylcholines/metabolism , Triglycerides/biosynthesis
11.
Cell Metab ; 25(6): 1282-1293.e7, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28416194

ABSTRACT

Immune cells function in diverse metabolic environments. Tissues with low glucose and high lactate concentrations, such as the intestinal tract or ischemic tissues, frequently require immune responses to be more pro-tolerant, avoiding unwanted reactions against self-antigens or commensal bacteria. T-regulatory cells (Tregs) maintain peripheral tolerance, but how Tregs function in low-glucose, lactate-rich environments is unknown. We report that the Treg transcription factor Foxp3 reprograms T cell metabolism by suppressing Myc and glycolysis, enhancing oxidative phosphorylation, and increasing nicotinamide adenine dinucleotide oxidation. These adaptations allow Tregs a metabolic advantage in low-glucose, lactate-rich environments; they resist lactate-mediated suppression of T cell function and proliferation. This metabolic phenotype may explain how Tregs promote peripheral immune tolerance during tissue injury but also how cancer cells evade immune destruction in the tumor microenvironment. Understanding Treg metabolism may therefore lead to novel approaches for selective immune modulation in cancer and autoimmune diseases.


Subject(s)
Cellular Microenvironment/immunology , Cellular Reprogramming/immunology , Forkhead Transcription Factors/immunology , Glucose/immunology , Lactic Acid/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Cellular Microenvironment/genetics , Cellular Reprogramming/genetics , Forkhead Transcription Factors/genetics , Glucose/genetics , Glycolysis/genetics , Glycolysis/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Oxidative Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology
12.
Cell Rep ; 18(8): 1906-1916, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228257

ABSTRACT

Long-lived plasma cells (PCs) in the bone marrow (BM) are a critical source of antibodies after infection or vaccination, but questions remain about the factors that control PCs. We found that systemic infection alters the BM, greatly reducing PCs and regulatory T (Treg) cells, a population that contributes to immune privilege in the BM. The use of intravital imaging revealed that BM Treg cells display a distinct behavior characterized by sustained co-localization with PCs and CD11c-YFP+ cells. Gene expression profiling indicated that BM Treg cells express high levels of Treg effector molecules, and CTLA-4 deletion in these cells resulted in elevated PCs. Furthermore, preservation of Treg cells during systemic infection prevents PC loss, while Treg cell depletion in uninfected mice reduced PC populations. These studies suggest a role for Treg cells in PC biology and provide a potential target for the modulation of PCs during vaccine-induced humoral responses or autoimmunity.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow/immunology , Plasma Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , CTLA-4 Antigen/immunology , Immunity, Humoral , Immunophenotyping/methods , Mice , Mice, Inbred C57BL
13.
Cell Metab ; 24(2): 269-82, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27508874

ABSTRACT

NAD is an obligate co-factor for the catabolism of metabolic fuels in all cell types. However, the availability of NAD in several tissues can become limited during genotoxic stress and the course of natural aging. The point at which NAD restriction imposes functional limitations on tissue physiology remains unknown. We examined this question in murine skeletal muscle by specifically depleting Nampt, an essential enzyme in the NAD salvage pathway. Knockout mice exhibited a dramatic 85% decline in intramuscular NAD content, accompanied by fiber degeneration and progressive loss of both muscle strength and treadmill endurance. Administration of the NAD precursor nicotinamide riboside rapidly ameliorated functional deficits and restored muscle mass despite having only a modest effect on the intramuscular NAD pool. Additionally, lifelong overexpression of Nampt preserved muscle NAD levels and exercise capacity in aged mice, supporting a critical role for tissue-autonomous NAD homeostasis in maintaining muscle mass and function.


Subject(s)
Homeostasis , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , NAD/metabolism , Administration, Oral , Aging/physiology , Animals , Biological Availability , Energy Metabolism , Glucose/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Muscle Strength , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiopathology , Necrosis , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Niacinamide/pharmacology , Nicotinamide Phosphoribosyltransferase/deficiency , Nicotinamide Phosphoribosyltransferase/metabolism , Organ Size , Physical Conditioning, Animal , Pyridinium Compounds , Transcription, Genetic
14.
Cell Metab ; 23(6): 1154-1166, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27238637

ABSTRACT

During insulin-resistant states such as type II diabetes mellitus (T2DM), insulin fails to suppress hepatic glucose production (HGP) yet promotes lipid synthesis. This metabolic state has been termed "selective insulin resistance" to indicate a defect in one arm of the insulin-signaling cascade, potentially downstream of Akt. Here we demonstrate that Akt-dependent activation of mTORC1 and inhibition of Foxo1 are required and sufficient for de novo lipogenesis, suggesting that hepatic insulin signaling is likely to be intact in insulin-resistant states. Moreover, cell-nonautonomous suppression of HGP by insulin depends on a reduction of adipocyte lipolysis and serum FFAs but is independent of vagal efferents or glucagon signaling. These data are consistent with a model in which, during T2DM, intact liver insulin signaling drives enhanced lipogenesis while excess circulating FFAs become a dominant inducer of nonsuppressible HGP.


Subject(s)
Glucose/biosynthesis , Hepatocytes/metabolism , Insulin/metabolism , Lipogenesis , Signal Transduction , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Diet , Efferent Pathways/drug effects , Efferent Pathways/metabolism , Fatty Acids, Nonesterified/metabolism , Forkhead Box Protein O1/metabolism , Gene Deletion , Gene Expression Regulation/drug effects , Glucagon/metabolism , Glucokinase/metabolism , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Glucose Tolerance Test , Heparin/pharmacology , Hepatocytes/drug effects , Insulin/pharmacology , Insulin Resistance , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/drug effects , Liver/innervation , Liver/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, Knockout , Multiprotein Complexes/metabolism , Postprandial Period/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Vagus Nerve/drug effects , Vagus Nerve/physiology
15.
PLoS One ; 10(6): e0126240, 2015.
Article in English | MEDLINE | ID: mdl-26042770

ABSTRACT

In non-alcoholic fatty liver disease (NAFLD) and insulin resistance, hepatic de novo lipogenesis is often elevated, but the underlying mechanisms remain poorly understood. Recently, we show that CDK8 functions to suppress de novo lipogenesis. Here, we identify the mammalian target of rapamycin complex 1 (mTORC1) as a critical regulator of CDK8 and its activating partner CycC. Using pharmacologic and genetic approaches, we show that increased mTORC1 activation causes the reduction of the CDK8-CycC complex in vitro and in mouse liver in vivo. In addition, mTORC1 is more active in three mouse models of NAFLD, correlated with the lower abundance of the CDK8-CycC complex. Consistent with the inhibitory role of CDK8 on de novo lipogenesis, nuclear SREBP-1c proteins and lipogenic enzymes are accumulated in NAFLD models. Thus, our results suggest that mTORC1 activation in NAFLD and insulin resistance results in down-regulation of the CDK8-CycC complex and elevation of lipogenic protein expression.


Subject(s)
Cyclin C/biosynthesis , Cyclin-Dependent Kinase 8/biosynthesis , Down-Regulation , Gene Expression Regulation, Enzymologic , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cyclin C/genetics , Cyclin-Dependent Kinase 8/genetics , HEK293 Cells , Humans , Lipogenesis/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Obese , Multiprotein Complexes/genetics , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , TOR Serine-Threonine Kinases/genetics
16.
Arthritis Rheum ; 65(4): 1043-54, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23334904

ABSTRACT

OBJECTIVE: To determine the necessity for any individual BAFF receptor in the development of systemic lupus erythematosus (SLE). METHODS: Bcma-, Taci-, and Br3-null mutations were introgressed into NZM 2328 mice. NZM.Bcma-/-, NZM.Taci-/-, and NZM.Br3-/- mice were evaluated for lymphocyte phenotype and BAFF receptor expression by flow cytometry; for B cell responsiveness to BAFF by in vitro culture; for serum levels of BAFF and total IgG and IgG anti-double-stranded DNA (anti-dsDNA) by enzyme-linked immunosorbent assay; for renal immunopathology by immunofluorescence and histopathology; and for clinical disease. RESULTS: BCMA, TACI, and B lymphocyte stimulator receptor 3 (BR3) were not surface-expressed in NZM.Bcma-/-, NZM.Taci-/-, and NZM.Br3-/- mice, respectively. Transitional and follicular B cells from NZM.Br3-/- mice were much less responsive to BAFF than were the corresponding cells from wild-type, NZM.Bcma-/-, or NZM.Taci-/- mice. In comparison with wild-type mice, NZM.Bcma-/- and NZM.Taci-/- mice harbored an increased number of spleen B cells, T cells, and plasma cells, whereas serum levels of total IgG and IgG anti-dsDNA were similar to those in wild-type mice. Despite their paucity of B cells, NZM.Br3-/- mice had an increased number of T cells, and the numbers of plasma cells and levels of IgG anti-dsDNA were similar to those in wild-type mice. Serum levels of BAFF were increased in NZM.Taci-/- and NZM.Br3-/- mice but were decreased in NZM.Bcma-/- mice. Despite their phenotypic differences, NZM.Bcma-/-, NZM.Taci-/-, and NZM.Br3-/- mice had renal immunopathology and clinical disease that were at least as severe as that in wild-type mice. CONCLUSION: Any single BAFF receptor, including BR3, is dispensable for the development of SLE in NZM mice. Development of disease in NZM.Br3-/- mice demonstrates that BAFF-BCMA and/or BAFF-TACI interactions contribute to SLE, and that a profound, life-long reduction in the numbers of B cells does not guarantee protection against SLE.


Subject(s)
B-Cell Maturation Antigen/metabolism , B-Lymphocyte Subsets , Lupus Erythematosus, Systemic/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , Animals , Antibodies, Antinuclear , B-Cell Activating Factor/pharmacology , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/metabolism , B-Cell Maturation Antigen/genetics , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Congenic , Transmembrane Activator and CAML Interactor Protein/genetics
18.
J Immunol ; 188(11): 5389-96, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22529295

ABSTRACT

The signals required to generate long-lived plasma cells remain unresolved. One widely cited model posits that long-lived plasma cells derive from germinal centers (GCs) in response to T cell-dependent (TD) Ags. Thus, T cell-independent (TI) Ags, which fail to sustain GCs, are considered ineffective at generating long-lived plasma cells. However, we show that long-lived hapten-specific plasma cells are readily induced without formation of GCs. Long-lived plasma cells developed in T cell-deficient mice after a single immunization with haptenated LPS, a widely used TI Ag. Long-lived plasma cells also formed in response to TD Ag when the GC response was experimentally prevented. These observations establish that long-lived plasma cells are induced in both TI and TD responses, and can arise independently of B cell maturation in GCs.


Subject(s)
Bone Marrow Cells/immunology , Cell Differentiation/immunology , Epitopes, T-Lymphocyte/immunology , Plasma Cells/immunology , T-Lymphocyte Subsets/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Lineage/immunology , Cell Survival/immunology , Epitopes, T-Lymphocyte/metabolism , Female , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plasma Cells/cytology , Plasma Cells/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
19.
Arthritis Rheum ; 64(5): 1610-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22127792

ABSTRACT

OBJECTIVE: To determine the role of APRIL in the development of systemic lupus erythematosus (SLE) in mice. METHODS: Wild-type (WT) NZM 2328, NZM. April(-/-) , NZM.Baff(-/-) , and NZM.Baff(-/-) .April(-/-) mice were evaluated for lymphocyte phenotype by flow cytometry, for serum total IgG and IgG autoantibody levels by enzyme-linked immunosorbent assay, for glomerular deposition of IgG and C3 by immunofluorescence, for renal changes by histopathology, and for clinical disease by laboratory assessment (severe proteinuria). RESULTS: In comparison to WT mice, NZM.April(-/-) mice harbored increased spleen B cells, T cells, and plasma cells (PCs), increased serum levels of IgG antichromatin antibodies, and decreased numbers of bone marrow (BM) PCs. Glomerular deposition of IgG and C3 was similar in NZM.April(-/-) mice and WT mice, renal changes on histopathology tended to be more severe in NZM.April(-/-) mice than in WT mice, and development of clinical disease was identical in NZM.April(-/-) mice and WT mice. BM (but not spleen) PCs and serum IgG antichromatin and anti-double-stranded DNA antibody levels were lower in NZM.Baff(-/-) .April(-/-) mice than in NZM.Baff(-/-) mice, whereas renal immunopathology in each cohort was equally mild. CONCLUSION: APRIL is dispensable for the development of full-blown SLE in NZM mice. Moreover, the elimination of both APRIL and BAFF had no discernible effect on the development of renal immunopathology or clinical disease beyond that of elimination of BAFF alone. The reduction in BM PCs in hosts doubly deficient in APRIL and BAFF beyond that in hosts deficient only in BAFF raises concern that combined antagonism of APRIL and BAFF may lead to greater immunosuppression without a concomitant increase in therapeutic efficacy.


Subject(s)
B-Cell Activating Factor/deficiency , Lupus Erythematosus, Systemic/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , B-Cell Activating Factor/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Biomarkers/metabolism , Bone Marrow Cells , Complement C3/immunology , Complement C3/metabolism , Disease Models, Animal , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunosuppression Therapy , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred NZB , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism , Plasma Cells/pathology , Species Specificity , Spleen/immunology , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
20.
J Clin Invest ; 121(10): 3954-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21881204

ABSTRACT

Antiviral Abs, for example those produced in response to influenza virus infection, are critical for virus neutralization and defense against secondary infection. While the half-life of Abs is short, Ab titers can last a lifetime due to a subset of the Ab-secreting cells (ASCs) that is long lived. However, the mechanisms governing ASC longevity are poorly understood. Here, we have identified a critical role for extrinsic cytokine signals in the survival of respiratory tract ASCs in a mouse model of influenza infection. Irradiation of mice at various time points after influenza virus infection markedly diminished numbers of lung ASCs, suggesting that they are short-lived and require extrinsic factors in order to persist. Neutralization of the TNF superfamily cytokines B lymphocyte stimulator (BLyS; also known as BAFF) and a proliferation-inducing ligand (APRIL) reduced numbers of antiviral ASCs in the lungs and bone marrow, whereas ASCs in the spleen and lung-draining lymph node were surprisingly unaffected. Mice deficient in transmembrane activator and calcium-modulator and cyclophilin ligand interactor (TACI), a receptor for BLyS and APRIL, mounted an initial antiviral B cell response similar to that generated in WT mice but failed to sustain protective Ab titers in the airways and serum, leading to increased susceptibility to secondary viral challenge. These studies highlight the importance of TACI signaling for the maintenance of ASCs and protection against influenza virus infection.


Subject(s)
Antibodies, Viral/biosynthesis , Orthomyxoviridae Infections/immunology , Transmembrane Activator and CAML Interactor Protein/immunology , Animals , Antibody-Producing Cells/immunology , Antibody-Producing Cells/pathology , Antibody-Producing Cells/radiation effects , B-Cell Activating Factor/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Survival , Disease Models, Animal , Female , Lung/immunology , Lung/pathology , Lung/radiation effects , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , Transmembrane Activator and CAML Interactor Protein/deficiency , Transmembrane Activator and CAML Interactor Protein/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...