Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Commun ; 15(1): 2598, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519468

ABSTRACT

Activation of the mechanistic target of rapamycin (mTOR) is a key metabolic checkpoint of pro-inflammatory T-cell development that contributes to the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE), however, the underlying mechanisms remain poorly understood. Here, we identify a functional role for Rab4A-directed endosome traffic in CD98 receptor recycling, mTOR activation, and accumulation of mitochondria that connect metabolic pathways with immune cell lineage development and lupus pathogenesis. Based on integrated analyses of gene expression, receptor traffic, and stable isotope tracing of metabolic pathways, constitutively active Rab4AQ72L exerts cell type-specific control over metabolic networks, dominantly impacting CD98-dependent kynurenine production, mTOR activation, mitochondrial electron transport and flux through the tricarboxylic acid cycle and thus expands CD4+ and CD3+CD4-CD8- double-negative T cells over CD8+ T cells, enhancing B cell activation, plasma cell development, antinuclear and antiphospholipid autoantibody production, and glomerulonephritis in lupus-prone mice. Rab4A deletion in T cells and pharmacological mTOR blockade restrain CD98 expression, mitochondrial metabolism and lineage skewing and attenuate glomerulonephritis. This study identifies Rab4A-directed endosome traffic as a multilevel regulator of T cell lineage specification during lupus pathogenesis.


Subject(s)
Glomerulonephritis , Lupus Erythematosus, Systemic , Animals , Mice , CD8-Positive T-Lymphocytes/metabolism , Endosomes/metabolism , Glomerulonephritis/metabolism , Kynurenine/metabolism , Mitochondria/metabolism , Mitophagy , TOR Serine-Threonine Kinases/metabolism , rab4 GTP-Binding Proteins/metabolism
3.
J Investig Med High Impact Case Rep ; 10: 23247096221127114, 2022.
Article in English | MEDLINE | ID: mdl-36341907

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare aggressive malignancy with poor outcomes. Although novel options like tagraxofusp, a CD123-directed cytotoxin, has emerged and is promising, treatment options are very limited in the relapsed and recurrent setting. We present a case of refractory BPDCN in a 62-year-old man who showed a complete bone marrow response to liposomal daunorubicin and cytarabine (vyxeos).


Subject(s)
Hematologic Neoplasms , Myeloproliferative Disorders , Skin Neoplasms , Male , Humans , Middle Aged , Hematologic Neoplasms/pathology , Cytarabine , Dendritic Cells/pathology , Skin Neoplasms/pathology , Daunorubicin , Acute Disease
4.
Clin Immunol Commun ; 1: 1-3, 2021 Dec.
Article in English | MEDLINE | ID: mdl-38620644

ABSTRACT

We describe a 64-year-old Caucasian female with a history of Raynaud's disease, hand arthritis, photosensitivity, Sjogren's syndrome and leukocytoclastic vasculitis who presented with progressively worsening fingertip necrosis that began three days after receiving a first dose of Pfizer-BioNTech COVID-19 RNA vaccine. Our workup revealed cryoglobulinemia, hypocomplementemia, elevated antinuclear antibodies (ANA) and IgM antiphospholipid autoantibodies (aPL) directed against phosphatidylserine (aPL-PS), suggesting a diagnosis of systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS). The patient failed to develop anti-spike IgG antibodies up to two months following vaccination. Disease progression was halted by plasmapheresis, anticoagulation, and immune suppression. We conclude that the vaccine RNA moiety may induce SLE manifesting in APS, cryoglobulinemia, hypocomplementemia, and digital necrosis.

5.
Ann Transl Med ; 8(14): 887, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32793731

ABSTRACT

Here, we present a 22-year-old female patient with adult-onset Still's disease (AOSD) who was newly diagnosed in the setting of secondary macrophage activation syndrome (MAS), a rare, life-threatening inflammatory disease with 50% mortality due to multi-organ failure. She met the diagnostic criteria of AOSD and MAS, while genetic testing excluded primary causes of MAS. She had high fevers, anemia, thrombocytopenia, splenomegaly, hematophagocytosis, and elevated serum ferritin (37,950 ng/mL) and CD25 levels (11,870 pg/mL), which remained unresponsive to corticosteroids and anakinra. Her serum interferon gamma (IFN-γ) levels were elevated (7 pg/mL). She was markedly responsive to IFN-γ blockade with emapalumab that eliminated her fevers and all MAS-associated laboratory abnormalities. This report provides initial evidence for therapeutic efficacy for IFN-γ blockade in AOSD and secondary MAS.

6.
Clin Immunol ; 214: 108388, 2020 05.
Article in English | MEDLINE | ID: mdl-32200114

ABSTRACT

Anti-phospholipid antibodies (aPL) and lupus anticoagulant (LAC) represent diagnostic criteria for systemic lupus erythematosus (SLE) and underlie anti-phospholipid syndrome (APS) in patients with and without SLE. 526 healthy controls and 1633 SLE and 1835 primary APS (PAPS) patients were evaluated. LAC was assessed by hexagonal phase phospholipid neutralization assay (HPPNA), diluted Russell viper venom test (dRVVT), and platelet neutralization procedure (PNP). ß2-glycoprotein-I and cardiolipin IgG, IgM, and IgA antibodies (aCL-IgG, aCL-IgM, aCL-IgA) were measured. 222/1633 SLE patients had APS based on the nine-test panel, which afforded the highest sensitivity (74%) and negative predictive value (90%) but lowest specificity (52%). HPPNA was the most sensitive individual test at 52%. The nine-test panel yielded the greatest sensitivity for aPL detection (70%) relative to HPPNA, the most sensitive individual test (36%) in PAPS. Superior sensitivity of a nine-test aPL panel has major implications for preventing potentially fatal thrombotic events in SLE and PAPS.


Subject(s)
Antiphospholipid Syndrome/diagnosis , Lupus Erythematosus, Systemic/complications , Antibodies, Antiphospholipid/blood , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/etiology , Humans , Lupus Coagulation Inhibitor/blood , Lupus Erythematosus, Systemic/blood , Platelet Function Tests , Predictive Value of Tests , Prothrombin Time , Retrospective Studies , Sensitivity and Specificity , Thrombosis/prevention & control , beta 2-Glycoprotein I/blood
7.
JCI Insight ; 5(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31805010

ABSTRACT

Overexpression and long terminal repeat (LTR) polymorphism of the HRES­1/Rab4 human endogenous retrovirus locus have been associated with T cell activation and disease manifestations in systemic lupus erythematosus (SLE). Although genomic DNA methylation is diminished overall in SLE, its role in HRES-1/Rab4 expression is unknown. Therefore, we determined how lupus-associated polymorphic rs451401 alleles of the LTR regulate transcription from the HRES-1/Rab4 promoter and thus affect T cell activation. The results showed that cytosine-119 is hypermethylated while cytosine-51 of the promoter and the LTR enhancer are hypomethylated in SLE. Pharmacologic or genetic inactivation of DNA methyltransferase 1 augmented the expression of HRES-1/Rab4. The minimal promoter was selectively recognized by metabolic stress sensor NRF1 when cytosine-119 but not cytosine-51 was methylated, and NRF1 stimulated HRES-1/Rab4 expression in human T cells. In turn, IRF2 and PSIP1 bound to the LTR enhancer and exerted control over HRES-1/Rab4 expression in rs451401 genotype- and methylation-dependent manners. The LTR enhancer conferred markedly greater expression of HRES-1/Rab4 in subjects with rs451401CC over rs451401GG alleles that in turn promoted mechanistic target of rapamycin (mTOR) activation upon T cell receptor stimulation. HRES-1/Rab4 alone robustly activated mTOR in human T cells. These findings identify HRES-1/Rab4 as a methylation- and rs451401 allele-dependent transducer of environmental stress and controller of T cell activation.


Subject(s)
Endogenous Retroviruses/genetics , Epigenesis, Genetic , Lupus Erythematosus, Systemic/genetics , TOR Serine-Threonine Kinases/genetics , Terminal Repeat Sequences/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Alleles , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Female , HCT116 Cells , HeLa Cells , Humans , Middle Aged , Nuclear Respiratory Factor 1 , Receptors, Antigen, T-Cell , T-Lymphocytes , Terminal Repeat Sequences/physiology , Transcription Factors , Young Adult
8.
J Inherit Metab Dis ; 43(3): 496-506, 2020 05.
Article in English | MEDLINE | ID: mdl-31769880

ABSTRACT

Transaldolase (TAL) is an enzyme in the pentose phosphate pathway (PPP) that generates NADPH for protection against oxidative stress. While deficiency of other PPP enzymes, such as transketolase (TKT), are incompatible with mammalian cell survival, mice lacking TAL are viable and develop progressive liver disease attributed to oxidative stress. Mice with homozygous or heterozygous TAL deficiency are predisposed to cirrhosis, hepatocellular carcinoma (HCC) and acetaminophen (APAP)-induced liver failure. Both mice and humans with complete TAL deficiency accumulate sedoheptulose 7-phosphate (S7P). Previous human studies relied on screening patients with S7P accumulation, thus excluding potentially pathogenic haploinsufficiency. Of note, mice with TAL haploinsufficiency are also predisposed to HCC and APAP-induced liver failure which are preventable with oral N-acetylcysteine (NAC) administration. Based on TALDO1 DNA sequencing, we detected functional TAL deficiency due to novel, heterozygous variations in two of 94 healthy adults and four of 27 subjects with APAP-induced liver failure (P = .022). The functional consequences of these variations were individually validated by site-directed mutagenesis of normal cDNA and loss of activity by recombinant enzyme. All four patients with TAL haplo-insufficiency with APAP-induced liver failure were successfully treated with NAC. We also document two novel variations in two of 15 children with previously unexplained liver cirrhosis. Examination of the National Center for Biotechnology Information databases revealed 274 coding region variations have been documented in 1125 TALDO1 sequences relative to 25 variations in 2870 TKT sequences (P < .0001). These findings suggest an unexpected prevalence and variety of genetic changes in human TALDO1 with relevance for liver injury that may be preventable by treatment with NAC.


Subject(s)
Acetylcysteine/pharmacology , Haploinsufficiency/drug effects , Liver Failure/chemically induced , Transaldolase/deficiency , Adult , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Female , Humans , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidative Stress/drug effects , Pentose Phosphate Pathway , Transaldolase/metabolism , Young Adult
9.
Clin Immunol ; 194: 100-104, 2018 09.
Article in English | MEDLINE | ID: mdl-30025818

ABSTRACT

In this study, we investigated four patients who met the diagnostic criteria for overlapping systemic lupus erythematosus (SLE) and myasthenia gravis (MG) but responded differently to treatment. All patients were acetylcholine receptor (AChR) and antinuclear antibody positive at the time of SLE diagnosis. Two patients presented with SLE who have been effectively treated with cholinesterase inhibitors for MG. These patients developed SLE with photosensitivity, rash, and arthritis post thymectomy, which had been performed 29 years and 40 years earlier, respectively. Two other patients were found to have AChR antibodies and MG in the context on new-onset SLE. These subjects were responsive to hydroxychloroquine and immunosuppression but failed cholinesterase inhibitors. The evolution of these cases is relevant for the role of thymus in lupus pathogenesis during aging and for treatment selection in SLE-MG overlap patients.


Subject(s)
Lupus Erythematosus, Systemic/diagnosis , Myasthenia Gravis/diagnosis , Female , Humans , Male , Middle Aged , Thymectomy/methods , Undifferentiated Connective Tissue Diseases/diagnosis
10.
Curr Rheumatol Rep ; 18(12): 73, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27812954

ABSTRACT

The mechanistic target of rapamycin (mTOR) is a central regulator in cell growth, activation, proliferation, and survival. Activation of the mTOR pathway underlies the pathogenesis of systemic lupus erythematosus (SLE). While mTOR activation and its therapeutic reversal were originally discovered in T cells, recent investigations have also uncovered roles in other cell subsets including B cells, macrophages, and "non-immune" organs such as the liver and the kidney. Activation of mTOR complex 1 (mTORC1) precedes the onset of SLE and associated co-morbidities, such as anti-phospholipid syndrome (APS), and may act as an early marker of disease pathogenesis. Six case reports have now been published that document the development of SLE in patients with genetic activation of mTORC1. Targeting mTORC1 over-activation with N-acetylcysteine, rapamycin, and rapalogs provides an opportunity to supplant current therapies with severe side effect profiles such as prednisone or cyclophosphamide. In the present review, we will discuss the recent explosion of findings in support for a central role for mTOR activation in SLE.


Subject(s)
B-Lymphocytes/metabolism , Lupus Erythematosus, Systemic/metabolism , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Acetylcysteine/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Sirolimus/therapeutic use
11.
Arthritis Rheumatol ; 68(11): 2728-2739, 2016 11.
Article in English | MEDLINE | ID: mdl-27332042

ABSTRACT

OBJECTIVE: Antiphospholipid antibodies (aPL) constitute a diagnostic criterion of systemic lupus erythematosus (SLE), and aPL have been functionally linked to liver disease in patients with SLE. Since the mechanistic target of rapamycin (mTOR) is a regulator of oxidative stress, a pathophysiologic process that contributes to the development of aPL, this study was undertaken in a mouse model of SLE to examine the involvement of liver mitochondria in lupus pathogenesis. METHODS: Mitochondria were isolated from lupus-prone MRL/lpr, C57BL/6.lpr, and MRL mice, age-matched autoimmunity-resistant C57BL/6 mice as negative controls, and transaldolase-deficient mice, a strain that exhibits oxidative stress in the liver. Electron transport chain (ETC) activity was assessed using measurements of oxygen consumption. ETC proteins, which are regulators of mitochondrial homeostasis, and the mTOR complexes mTORC1 and mTORC2 were examined by Western blotting. Anticardiolipin (aCL) and anti-ß2 -glycoprotein I (anti-ß2 GPI) autoantibodies were measured by enzyme-linked immunosorbent assay in mice treated with rapamycin or mice treated with a solvent control. RESULTS: Mitochondrial oxygen consumption was increased in the livers of 4-week-old, disease-free MRL/lpr mice relative to age-matched controls. Levels of the mitophagy initiator dynamin-related protein 1 (Drp1) were depleted while the activity of mTORC1 was increased in MRL/lpr mice. In turn, mTORC2 activity was decreased in MRL and MRL/lpr mice. In addition, levels of aCL and anti-ß2 GPI were elevated preceding the development of nephritis in 4-week-old MRL, C57BL/6.lpr, and MRL/lpr mice. Transaldolase-deficient mice showed increased oxygen consumption, depletion of Drp1, activation of mTORC1, and elevated expression of NADH:ubiquinone oxidoreductase core subunit S3 (NDUFS3), a pro-oxidant subunit of ETC complex I, as well as increased production of aCL and anti-ß2 GPI autoantibodies. Treatment with rapamycin selectively blocked mTORC1 activation, NDUFS3 expression, and aPL production both in transaldolase-deficient mice and in lupus-prone mice. CONCLUSION: In lupus-prone mice, mTORC1-dependent mitochondrial dysfunction contributes to the generation of aPL, suggesting that such mechanisms may represent a treatment target in patients with SLE.


Subject(s)
Antibodies, Antiphospholipid/biosynthesis , Electron Transport Chain Complex Proteins/metabolism , Lupus Erythematosus, Systemic/immunology , Mitochondria, Liver/metabolism , Multiprotein Complexes/metabolism , Oxidative Stress/immunology , Oxygen Consumption/immunology , TOR Serine-Threonine Kinases/metabolism , Animals , Antibodies, Anticardiolipin/biosynthesis , Antibodies, Anticardiolipin/drug effects , Antibodies, Anticardiolipin/immunology , Antibodies, Antiphospholipid/drug effects , Antibodies, Antiphospholipid/immunology , Antibody Formation/drug effects , Antibody Formation/immunology , Blotting, Western , Disease Models, Animal , Dynamins/metabolism , Electron Transport Chain Complex Proteins/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Immunosuppressive Agents/pharmacology , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Sirolimus/pharmacology , Transaldolase/genetics , beta 2-Glycoprotein I/immunology
12.
Clin Immunol ; 160(2): 319-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26160213

ABSTRACT

Liver disease (LD), defined as ≥ 2-fold elevation of aspartate aminotransferase (AST) or alanine aminotransferase (ALT), was examined in a longitudinal study of systemic lupus erythematosus (SLE) patients. Among 435 patients, 90 (20.7%) had LD with a greater prevalence in males (15/39; 38.5%) than females (75/396; 18.9%; p = 0.01). SLE disease activity index (SLEDAI) was greater in LD patients (7.8 ± 0.7) relative to those without (5.8 ± 0.3; p = 0.0025). Anti-smooth muscle antibodies, anti-DNA antibodies, hypocomplementemia, proteinuria, leucopenia, thrombocytopenia, and anti-phospholipid syndrome were increased in LD. An absence of LD was noted in patients receiving rapamycin relative to azathioprine, cyclosporine A, or cyclophosphamide. An absence of LD was also noted in patients treated with N-acetylcysteine. LFTs were normalized and SLEDAI was diminished with increased prednisone use in 76/90 LD patients over 12.1 ± 2.6 months. Thus, LD is attributed to autoimmunity and disease activity, it responds to prednisone, and it is potentially preventable by rapamycin or N-acetylcysteine treatment.


Subject(s)
Antibodies, Antinuclear/immunology , Liver Diseases/immunology , Lupus Erythematosus, Systemic/immunology , Acetylcysteine/therapeutic use , Adult , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Azathioprine/therapeutic use , Biomarkers , Cohort Studies , Complement System Proteins/immunology , Cyclosporine/therapeutic use , Diabetes Mellitus/epidemiology , Female , Free Radical Scavengers/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Liver Diseases/drug therapy , Liver Diseases/epidemiology , Longitudinal Studies , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/epidemiology , Male , Middle Aged , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Prednisone/therapeutic use , Prevalence , Retrospective Studies , Severity of Illness Index , Sex Distribution , Sirolimus/therapeutic use
13.
PLoS One ; 9(1): e84392, 2014.
Article in English | MEDLINE | ID: mdl-24404161

ABSTRACT

HRES-1/Rab4 is a small GTPase that regulates endocytic recycling. It has been colocalized to mitochondria and the mechanistic target of rapamycin (mTOR), a suppressor of autophagy. Since the autophagosomal membrane component microtubule-associated protein light chain 3 (LC3) is derived from mitochondria, we investigated the impact of HRES-1/Rab4 on the formation of LC3(+) autophagosomes, their colocalization with HRES-1/Rab4 and mitochondria, and the retention of mitochondria during autophagy induced by starvation and rapamycin. HRES-1/Rab4 exhibited minimal baseline colocalization with LC3, which was enhanced 22-fold upon starvation or 6-fold upon rapamycin treatment. Colocalization of HRES-1/Rab4 with mitochondria was increased >2-fold by starvation or rapamycin. HRES-1/Rab4 overexpression promoted the colocalization of mitochondria with LC3 upon starvation or rapamycin treatment. A dominant-negative mutant, HRES-1/Rab4(S27N) had reduced colocalization with LC3 and mitochondria upon starvation but not rapamycin treatment. A constitutively active mutant, HRES-1/Rab4(Q72L) showed diminished colocalization with LC3 but promoted the partitioning of mitochondria with LC3 upon starvation or rapamycin treatment. Phosphorylation-resistant mutant HRES-1/Rab4(S204Q) showed diminished colocalization with LC3 but increased partitioning to mitochondria. A newly discovered C-terminally truncated native isoform, HRES-1/Rab4(1-121), showed enhanced localization to LC3 and mitochondria without starvation or rapamycin treatment. HRES-1/Rab4(1-121) increased the formation of LC3(+) autophagosomes in resting cells, while other isoforms promoted autophagosome formation upon starvation. HRES-1/Rab4, HRES-1/Rab4(1-121), HRES-1/Rab4(Q72L) and HRES-1/Rab4(S204Q) promoted the accumulation of mitochondria during starvation. The specificity of HRES-1/Rab4-mediated mitochondrial accumulation is indicated by its abrogation by dominant-negative HRES-1/Rab4(S27N) mutation. The formation of interconnected mitochondrial tubular networks was markedly enhanced by HRES-1/Rab4(Q72L) upon starvation, which may contribute to the retention of mitochondria during autophagy. The present study thus indicates that HRES-1/Rab4 regulates autophagy through promoting the formation of LC3(+) autophagosomes and the preservation of mitochondria.


Subject(s)
Autophagy/physiology , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Phagosomes/metabolism , rab4 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Cell Line , Gene Expression , Genes, Reporter , Humans , Intracellular Membranes/metabolism , Macrolides/pharmacology , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Mitochondria/drug effects , Molecular Sequence Data , Protein Binding , Protein Transport/drug effects , rab4 GTP-Binding Proteins/genetics
14.
Ann Rheum Dis ; 73(10): 1888-97, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23897774

ABSTRACT

OBJECTIVE: Accumulation of mitochondria underlies T-cell dysfunction in systemic lupus erythematosus (SLE). Mitochondrial turnover involves endosomal traffic regulated by HRES-1/Rab4, a small GTPase that is overexpressed in lupus T cells. Therefore, we investigated whether (1) HRES-1/Rab4 impacts mitochondrial homeostasis and (2) Rab geranylgeranyl transferase inhibitor 3-PEHPC blocks mitochondrial accumulation in T cells, autoimmunity and disease development in lupus-prone mice. METHODS: Mitochondria were evaluated in peripheral blood lymphocytes (PBL) of 38 SLE patients and 21 healthy controls and mouse models by flow cytometry, microscopy and western blot. MRL/lpr mice were treated with 125 µg/kg 3-PEHPC or 1 mg/kg rapamycin for 10 weeks, from 4 weeks of age. Disease was monitored by antinuclear antibody (ANA) production, proteinuria, and renal histology. RESULTS: Overexpression of HRES-1/Rab4 increased the mitochondrial mass of PBL (1.4-fold; p=0.019) and Jurkat cells (2-fold; p=0.000016) and depleted the mitophagy initiator protein Drp1 both in human (-49%; p=0.01) and mouse lymphocytes (-41%; p=0.03). Drp1 protein levels were profoundly diminished in PBL of SLE patients (-86±3%; p=0.012). T cells of 4-week-old MRL/lpr mice exhibited 4.7-fold over-expression of Rab4A (p=0.0002), the murine homologue of HRES-1/Rab4, and depletion of Drp1 that preceded the accumulation of mitochondria, ANA production and nephritis. 3-PEHPC increased Drp1 (p=0.03) and reduced mitochondrial mass in T cells (p=0.02) and diminished ANA production (p=0.021), proteinuria (p=0.00004), and nephritis scores of lupus-prone mice (p<0.001). CONCLUSIONS: These data reveal a pathogenic role for HRES-1/Rab4-mediated Drp1 depletion and identify endocytic control of mitophagy as a treatment target in SLE.


Subject(s)
GTP Phosphohydrolases/blood , Lupus Erythematosus, Systemic/blood , Microtubule-Associated Proteins/blood , Mitochondria/metabolism , Mitochondrial Proteins/blood , rab4 GTP-Binding Proteins/physiology , Animals , Autophagy/physiology , Case-Control Studies , Cells, Cultured , Diphosphonates/therapeutic use , Dynamins/blood , Dynamins/physiology , Female , GTP Phosphohydrolases/physiology , Homeostasis/physiology , Humans , Jurkat Cells , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lysosomes/metabolism , Mice, Inbred MRL lpr , Microtubule-Associated Proteins/physiology , Mitochondrial Proteins/physiology , Mitophagy/immunology , Molecular Targeted Therapy/methods , Pyridines/therapeutic use , T-Lymphocytes/metabolism
16.
J Immunol ; 184(7): 4025-32, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20194725

ABSTRACT

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the CNS resulting from a progressive loss of oligodendrocytes. Transaldolase (TAL) is expressed at selectively high levels in oligodendrocytes of the brain, and postmortem sections show concurrent loss of myelin basic protein and TAL from sites of demyelination. Infiltrating CD8(+) CTLs are thought to play a key role in oligodendrocyte cell death. Cleavage by granzyme B (GrB) is predictive for autoantigenicity of self-proteins, thereby further implicating CTL-induced death in the initiation and propagation of autoimmunity. The precursor frequency and CTL activity of HLA-A2-restricted TAL 168-176-specific CD8(+) T cells is increased in MS patients. In this paper, we show that TAL, but not myelin basic protein, is specifically cleaved by human GrB. The recognition site of GrB that resulted in the cleavage of a dominant TAL fragment was mapped to a VVAD motif at aa residue 27 by N-terminal sequencing and confirmed by site-directed mutagenesis. The major C-terminal GrB cleavage product, residues 28-337, had no enzymatic activity but retained the antigenicity of full-length TAL, effectively stimulating the proliferation and CTL activity of PBMCs and of CD8(+) T cell lines from patients with MS. Sera of MS patients exhibited similar binding affinity to wild-type and GrB-cleaved TAL. Because GrB mediates the killing of target cells and cleavage by GrB is predictive of autoantigen status of self proteins, GrB-cleaved TAL-specific T cell-mediated cytotoxicity may contribute to the progressive destruction of oligodendrocytes in patients with MS.


Subject(s)
Autoantigens/immunology , Granzymes/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Transaldolase/immunology , Amino Acid Sequence , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/metabolism , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodendroglia/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transaldolase/metabolism
17.
J Clin Invest ; 119(6): 1546-57, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19436114

ABSTRACT

Although oxidative stress has been implicated in acute acetaminophen-induced liver failure and in chronic liver cirrhosis and hepatocellular carcinoma (HCC), no common underlying metabolic pathway has been identified. Recent case reports suggest a link between the pentose phosphate pathway (PPP) enzyme transaldolase (TAL; encoded by TALDO1) and liver failure in children. Here, we show that Taldo1-/- and Taldo1+/- mice spontaneously developed HCC, and Taldo1-/- mice had increased susceptibility to acetaminophen-induced liver failure. Oxidative stress in Taldo1-/- livers was characterized by the accumulation of sedoheptulose 7-phosphate, failure to recycle ribose 5-phosphate for the oxidative PPP, depleted NADPH and glutathione levels, and increased production of lipid hydroperoxides. Furthermore, we found evidence of hepatic mitochondrial dysfunction, as indicated by loss of transmembrane potential, diminished mitochondrial mass, and reduced ATP/ADP ratio. Reduced beta-catenin phosphorylation and enhanced c-Jun expression in Taldo1-/- livers reflected adaptation to oxidative stress. Taldo1-/- hepatocytes were resistant to CD95/Fas-mediated apoptosis in vitro and in vivo. Remarkably, lifelong administration of the potent antioxidant N-acetylcysteine (NAC) prevented acetaminophen-induced liver failure, restored Fas-dependent hepatocyte apoptosis, and blocked hepatocarcinogenesis in Taldo1-/- mice. These data reveal a protective role for the TAL-mediated branch of the PPP against hepatocarcinogenesis and identify NAC as a promising treatment for liver disease in TAL deficiency.


Subject(s)
Acetylcysteine/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/prevention & control , Cell Transformation, Neoplastic/metabolism , Liver Failure/chemically induced , Liver Neoplasms/enzymology , Transaldolase/deficiency , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Phosphorylation , Transaldolase/metabolism , alpha-Fetoproteins/metabolism , beta Catenin/metabolism , fas Receptor/metabolism
18.
Biochem J ; 415(1): 123-34, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18498245

ABSTRACT

TAL (transaldolase) was originally described in the yeast as an enzyme of the PPP (pentose phosphate pathway). However, certain organisms and mammalian tissues lack TAL, and the overall reason for its existence is unclear. Recently, deletion of Ser(171) (TALDeltaS171) was found in five patients causing inactivation, proteasome-mediated degradation and complete deficiency of TAL. In the present study, microarray and follow-up Western-blot, enzyme-activity and metabolic studies of TALDeltaS171 TD (TAL-deficient) lymphoblasts revealed co-ordinated changes in the expression of genes involved in the PPP, mitochondrial biogenesis, oxidative stress, and Ca(2+) fluxing. Sedoheptulose 7-phosphate was accumulated, whereas G6P (glucose 6-phosphate) was depleted, indicating a failure to recycle G6P for the oxidative branch of the PPP. Nucleotide analysis showed depletion of NADPH and NAD(+) and accumulation of ADP-ribose. TD cells have diminished Deltapsi(m) (mitochondrial transmembrane potential) and increased mitochondrial mass associated with increased production of nitric oxide and ATP. TAL deficiency resulted in enhanced spontaneous and H(2)O(2)-induced apoptosis. TD lymphoblasts showed increased expression of CD38, which hydrolyses NAD(+) into ADP-ribose, a trigger of Ca(2+) release from the endoplasmic reticulum that, in turn, facilitated CD20-induced apoptosis. By contrast, TD cells were resistant to CD95/Fas-induced apoptosis, owing to a dependence of caspase activity on redox-sensitive cysteine residues. Normalization of TAL activity by adeno-associated-virus-mediated gene transfer reversed the elevated CD38 expression, ATP and Ca(2+) levels, suppressed H(2)O(2)- and CD20-induced apoptosis and enhanced Fas-induced cell death. The present study identified the TAL deficiency as a modulator of mitochondrial homoeostasis, Ca(2+) fluxing and apoptosis.


Subject(s)
Apoptosis/physiology , Homeostasis/physiology , Mitochondria/physiology , Pentose Phosphate Pathway/physiology , Transaldolase/deficiency , Cell Line, Transformed , Cells, Cultured , Female , Glucose-6-Phosphate/metabolism , Humans , Microscopy, Electron , Signal Transduction , Sugar Phosphates/metabolism , Transaldolase/genetics
19.
Proc Natl Acad Sci U S A ; 103(40): 14813-8, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-17003133

ABSTRACT

Fertility of spermatozoa depends on maintenance of the mitochondrial transmembrane potential (Deltapsi(m)), which is generated by the electron-transport chain and regulated by an oxidation-reduction equilibrium of reactive oxygen intermediates, pyridine nucleotides, and glutathione (GSH). Here, we report that male mice lacking transaldolase (TAL)(-/-) are sterile because of defective forward motility. TAL(-/-) spermatozoa show loss of Deltapsi(m) and mitochondrial membrane integrity because of diminished NADPH, NADH, and GSH. Mitochondria constitute major Ca(2+) stores; thus, diminished mitochondrial mass accounts for reduced Ca(2+) fluxing, defective forward motility, and infertility. Reduced forward progression of TAL-deficient spermatozoa is associated with diminished mitochondrial reactive oxygen intermediate production and Ca(2+) levels, intracellular acidosis, and compensatory down-regulation of carbonic anhydrase IV and overexpression of CD38 and gamma-glutamyl transferase. Microarray analyses of gene expression in the testis, caput, and cauda epididymidis of TAL(+/+), TAL(+/-), and TAL(-/-) littermates confirmed a dominant impact of TAL deficiency on late stages of sperm-cell development, affecting the electron-transport chain and GSH metabolism. Stimulation of de novo GSH synthesis by oral N-acetyl-cysteine normalized the low fertility rate of TAL(+/-) males without affecting the sterility of TAL(-/-) males. Whereas TAL(-/-) sperm failed to fertilize TAL(+/+) oocytes in vitro, sterility of TAL(-/-) sperm was circumvented by intracytoplasmic sperm injection, indicating that TAL deficiency influenced the structure and function of mitochondria without compromising the nucleus and DNA integrity. Collectively, these data reveal an essential role of TAL in sperm-cell mitochondrial function and, thus, male fertility.


Subject(s)
Fertility/physiology , Mitochondrial Membranes/enzymology , Spermatozoa/enzymology , Spermatozoa/physiology , Transaldolase/metabolism , Animals , Calcium Signaling/physiology , Epididymis/enzymology , Epididymis/ultrastructure , Gene Expression , Gene Silencing , Heterozygote , Homozygote , Infertility, Male , Male , Membrane Potentials/physiology , Mice , Mice, Knockout , Mitochondria/pathology , Mitochondria/ultrastructure , Molecular Sequence Data , NAD/metabolism , Reactive Oxygen Species/metabolism , Recombination, Genetic , Sperm Motility/physiology , Spermatozoa/cytology , Spermatozoa/ultrastructure , Sugar Phosphates/metabolism , Transaldolase/deficiency
20.
J Biol Chem ; 281(45): 34574-91, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16935861

ABSTRACT

A novel 2986-base transcript encoded by the antisense strand of the HRES-1 human endogenous retrovirus was isolated from peripheral blood lymphocytes. This transcript codes for a 218-amino acid protein, termed HRES-1/Rab4, based on homology to the Rab4 family of small GTPases. Antibody 13407 raised against recombinant HRES-1/Rab4 detected a native protein of identical molecular weight in human T cells. HRES-1 nucleotides 2151-1606, located upstream of HRES-1/Rab4 exon 1, have promoter activity when oriented in the direction of HRES-1/Rab4 transcription. The human immunodeficiency virus, type 1 (HIV-1), tat gene stimulates transcriptional activity of the HRES-1/Rab4 promoter via trans-activation of the HRES-1 long terminal repeat. Transfection of HIV-1 tat into HeLa cells or infection of H9 and Jurkat cells by HIV-1 increased HRES-1/Rab4 protein levels. Overexpression of HRES-1/Rab4 in Jurkat cells abrogated HIV infection, gag p24 production, and apoptosis, whereas dominant-negative HRES-1/Rab4(S27N) had the opposite effects. HRES-1/Rab4 inhibited surface expression of CD4 and targeted it for lysosomal degradation. HRES-1/Rab4(S27N) enhanced surface expression, recycling, and total cellular CD4 content. Infection by HIV elicited a coordinate down-regulation of CD4 and up-regulation of HRES-1/Rab4 in PBL. Moreover, overexpression of HRES-1/Rab4 reduced CD4 expression on peripheral blood CD4+ T cells. Stimulation by HIV-1 of HRES-1/Rab4 expression and its regulation of CD4 recycling reveal novel coordinate interactions between an infectious retrovirus and the human genome.


Subject(s)
CD4 Antigens/metabolism , Gene Expression Regulation, Viral , Gene Products, tat/genetics , HIV Infections/metabolism , HIV-1/pathogenicity , rab4 GTP-Binding Proteins/metabolism , Antigens, CD/metabolism , Apoptosis , Base Sequence , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Chloramphenicol O-Acetyltransferase/metabolism , Dependovirus/genetics , Disease Susceptibility , Exons/genetics , Flow Cytometry , Gene Products, tat/pharmacology , Genes, Dominant , HIV Core Protein p24/metabolism , HIV Infections/virology , HIV Long Terminal Repeat/genetics , HeLa Cells , Humans , Introns/genetics , Jurkat Cells , Lysosomes , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , Transfection , rab4 GTP-Binding Proteins/genetics , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...