Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Sci Transl Med ; 12(551)2020 07 08.
Article in English | MEDLINE | ID: mdl-32641487

ABSTRACT

The autoimmune disease systemic lupus erythematosus (SLE) is characterized by the production of pathogenic autoantibodies. It has been postulated that gut microbial dysbiosis may be one of the mechanisms involved in SLE pathogenesis. Here, we demonstrate that the dysbiotic gut microbiota of triple congenic (TC) lupus-prone mice (B6.Sle1.Sle2.Sle3) stimulated the production of autoantibodies and activated immune cells when transferred into germfree congenic C57BL/6 (B6) mice. Fecal transfer to B6 mice induced autoimmune phenotypes only when the TC donor mice exhibited autoimmunity. Autoimmune pathogenesis was mitigated by horizontal transfer of the gut microbiota between co-housed lupus-prone TC mice and control congenic B6 mice. Metabolomic screening identified an altered distribution of tryptophan metabolites in the feces of TC mice including an increase in kynurenine, which was alleviated after antibiotic treatment. Low dietary tryptophan prevented autoimmune pathology in TC mice, whereas high dietary tryptophan exacerbated disease. Reducing dietary tryptophan altered gut microbial taxa in both lupus-prone TC mice and control B6 mice. Consequently, fecal transfer from TC mice fed a high tryptophan diet, but not a low tryptophan diet, induced autoimmune phenotypes in germfree B6 mice. The interplay of gut microbial dysbiosis, tryptophan metabolism and host genetic susceptibility in lupus-prone mice suggest that aberrant tryptophan metabolism may contribute to autoimmune activation in this disease.


Subject(s)
Gastrointestinal Microbiome , Lupus Erythematosus, Systemic , Animals , Autoimmunity , Dysbiosis , Mice , Mice, Inbred C57BL , Tryptophan
2.
J Immunol ; 197(2): 458-69, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27296664

ABSTRACT

Pbx1 controls chromatin accessibility to a large number of genes and is entirely conserved between mice and humans. The Pbx1-d dominant-negative isoform is more frequent in CD4(+) T cells from lupus patients than from healthy controls. Pbx1-d is associated with the production of autoreactive T cells in mice carrying the Sle1a1 lupus-susceptibility locus. Transgenic (Tg) expression of Pbx1-d in CD4(+) T cells reproduced the phenotypes of Sle1a1 mice, with increased inflammatory functions of CD4(+) T cells and impaired Foxp3(+) regulatory T cell (Treg) homeostasis. Pbx1-d-Tg expression also expanded the number of follicular helper T cells (TFHs) in a cell-intrinsic and Ag-specific manner, which was enhanced in recall responses and resulted in Th1-biased Abs. Moreover, Pbx1-d-Tg CD4(+) T cells upregulated the expression of miR-10a, miR-21, and miR-155, which were implicated in Treg and follicular helper T cell homeostasis. Our results suggest that Pbx1-d impacts lupus development by regulating effector T cell differentiation and promoting TFHs at the expense of Tregs. In addition, our results identify Pbx1 as a novel regulator of CD4(+) T cell effector function.


Subject(s)
Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Lupus Erythematosus, Systemic/immunology , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Flow Cytometry , Homeodomain Proteins/immunology , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Polymerase Chain Reaction , Pre-B-Cell Leukemia Transcription Factor 1 , Transcription Factors/immunology
3.
J Immunol ; 196(1): 80-90, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26608911

ABSTRACT

We have previously shown that CD4(+) T cells from B6.Sle1Sle2.Sle3 lupus mice and patients present a high cellular metabolism, and a treatment combining 2-deoxy-D-glucose, which inhibits glucose metabolism, and metformin, which inhibits oxygen consumption, normalized lupus T cell functions in vitro and reverted disease in mice. We obtained similar results with B6.lpr mice, another model of lupus, and showed that a continuous treatment is required to maintain the beneficial effect of metabolic inhibitors. Further, we investigated the relative roles of glucose oxidation and pyruvate reduction into lactate in this process. Treatments of B6.Sle1Sle2.Sle3 mice with either 2-deoxy-D-glucose or metformin were sufficient to prevent autoimmune activation, whereas their combination was necessary to reverse the process. Treatment of B6.Sle1Sle2.Sle3 mice with dichloroacetate, an inhibitor of lactate production, failed to effectively prevent or reverse autoimmune pathology. In vitro, CD4(+) T cell activation upregulated the expression of genes that favor oxidative phosphorylation. Blocking glucose oxidation inhibited both IFN-γ and IL-17 production, which could not be achieved by blocking pyruvate reduction. Overall, our data show that targeting glucose oxidation is required to prevent or reverse lupus development in mice, which cannot be achieved by simply targeting the pyruvate-lactate conversion.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Glucose/metabolism , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Oxidation-Reduction/drug effects , Animals , Autoimmunity/drug effects , Cells, Cultured , Deoxyglucose/pharmacology , Dichloroacetic Acid/pharmacology , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/immunology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Lactic Acid/biosynthesis , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Phosphorylation , Oxygen Consumption/drug effects , Oxygen Consumption/immunology , Pyruvic Acid/metabolism
4.
Arterioscler Thromb Vasc Biol ; 36(2): 266-73, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26634654

ABSTRACT

OBJECTIVE: Patients with systemic lupus erythematosis are at risk for premature atherosclerosis and half of the patients with systemic lupus erythematosis have elevated type I interferon (IFN-I) levels. We hypothesized that IFN-I would induce premature atherosclerosis by increasing the number of smooth muscle progenitor cells (SMPC) in the bloodstream and promoting atherosclerotic lesions within the vasculature. APPROACH AND RESULTS: SMPC isolated from wild-type and IFN receptor knockout animals were cultured in medium±IFN-I. In vivo, we used electroporation to generate stable IFN-I expression for as long as 4 months. The number of SMPC was determined in mice that expressed IFN-I and in control mice and sections from the bifurcation of the abdominal aorta were analyzed 3 months after electroporation of an IFN-I expression plasmid or a control plasmid. Adding IFN-I to the media increased the number of cultured wild-type SMPC and increased mRNA for SM22, but had no effect on SMPC isolated from IFN receptor knockout mice. Our in vivo results demonstrated a positive relationship between the preatherosclerotic-like lesions and endothelial damage. Although, there were no significant differences in smooth muscle cell density or thickness of the medial layer between groups, the IFN-I-expressing mice had a significant increase in preatherosclerotic-like lesions and immature smooth muscle cells, cells that expressed CD34 and smooth muscle α-actin; but lacked smooth muscle myosin heavy chain. CONCLUSIONS: IFN-I seems to enhance SMPC number in vitro. In vivo IFN-I expression may maintain SMPC in an immature state. These immature smooth muscle cells could give rise to macrophages and eventually foam cells.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cell Differentiation , Interferon Type I/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Stem Cells/metabolism , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Genotype , Interferon Type I/deficiency , Interferon Type I/genetics , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Myosin Heavy Chains/metabolism , Phenotype , Stem Cells/pathology , Time Factors , Transfection
5.
Sci Transl Med ; 7(274): 274ra18, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673763

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease in which autoreactive CD4(+) T cells play an essential role. CD4(+) T cells rely on glycolysis for inflammatory effector functions, but recent studies have shown that mitochondrial metabolism supports their chronic activation. How these processes contribute to lupus is unclear. We show that both glycolysis and mitochondrial oxidative metabolism are elevated in CD4(+) T cells from lupus-prone B6.Sle1.Sle2.Sle3 (TC) mice as compared to non-autoimmune controls. In vitro, both the mitochondrial metabolism inhibitor metformin and the glucose metabolism inhibitor 2-deoxy-d-glucose (2DG) reduced interferon-γ (IFN-γ) production, although at different stages of activation. Metformin also restored the defective interleukin-2 (IL-2) production by TC CD4(+) T cells. In vivo, treatment of TC mice and other lupus models with a combination of metformin and 2DG normalized T cell metabolism and reversed disease biomarkers. Further, CD4(+) T cells from SLE patients also exhibited enhanced glycolysis and mitochondrial metabolism that correlated with their activation status, and their excessive IFN-γ production was significantly reduced by metformin in vitro. These results suggest that normalization of T cell metabolism through the dual inhibition of glycolysis and mitochondrial metabolism is a promising therapeutic venue for SLE.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Lupus Erythematosus, Systemic/metabolism , Animals , Deoxyglucose/therapeutic use , Disease Models, Animal , Lupus Erythematosus, Systemic/drug therapy , Metformin/therapeutic use , Mice , Phenotype
6.
J Immunol ; 194(1): 43-55, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25416808

ABSTRACT

Pre­B cell leukemia homeobox 1 (Pbx1)-d is a dominant-negative splice isoform of the gene Pbx1 that corresponds to the NZM2410 lupus susceptibility locus Sle1a1. Pbx1 is required to maintain stem cell self-renewal, including that of mesenchymal stem cells (MSCs). MSCs have immunosuppressive functions that require stem cell maintenance. We tested the hypothesis that the expression of Pbx1-d favors MSC differentiation and impairs their immunosuppressive functions. We demonstrate that Sle1a1 MSCs express high levels of Pbx1-d as compared with congenic C57BL/6J (B6) MSCs. Sle1a1 MSCs grew faster and differentiated significantly more rapidly into osteoblasts than did B6 MSCs. This corresponded to a significant decrease in the expression of genes associated with stemness and an increase in the expression of genes associated with differentiation. Additionally, Sle1a1 MSCs express a gene expression profile associated with an enhanced innate immunity and inflammation. Suppression of Ig production from TLR-activated B6 B cells and IL-2 secretion from activated B6 CD4+ T cells was significantly impaired in Sle1a1 MSCs as compared with B6 MSCs. B6.Sle1a1 MSCs showed intermediate activity in suppressing lupus immunophenotypes in three different mouse models. Taken together, these data suggest that the expression of the lupus susceptibility allele Pbx1-d isoform impairs MSC functions, which may contribute to lupus pathogenesis both through a defective immunosuppression and the promotion of a proinflammatory environment.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Homeodomain Proteins/genetics , Immune Tolerance/immunology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Transcription Factors/genetics , Animals , Autoimmunity/genetics , Gene Expression , Homeodomain Proteins/biosynthesis , Immunoglobulins/biosynthesis , Immunologic Memory/immunology , Inflammation/immunology , Interleukin-2/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Pre-B-Cell Leukemia Transcription Factor 1 , Protein Isoforms/genetics , Toll-Like Receptors/immunology , Transcription Factors/biosynthesis
7.
Diabetes Care ; 36(11): 3815-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24041678

ABSTRACT

OBJECTIVE: Evidence supporting an association between complement (C) and type 1 diabetes (T1D) includes the identification of C-fixing islet cell autoantibodies in T1D sera and genetic associations with the major histocompatibility complex III C4 region on chromosome 6. Therefore, we investigated whether C activation was present in pancreata from those with or at increased risk (positive for T1D associated autoantibodies) for T1D. RESEARCH DESIGN AND METHODS: Immunohistochemical techniques were used to measure the C degradation product C4d in organ donor pancreata from patients with T1D and type 2 diabetes and autoantibody-positive and autoantibody-negative subjects. RESULTS: Median C4d antigen density differed across the groups (P < 0.0001) and was highest in patients with T1D. C4d immunostaining localized to the blood vessel endothelium and extracellular matrix surrounding blood vessels and exocrine ducts. Receiver operating characteristic analysis resulted in 81.8% sensitivity and 94.4% specificity for C4d staining. CONCLUSIONS: These data suggest that C activation is occurring within pancreata from patients with T1D and C4d may be a biomarker for T1D.


Subject(s)
Complement Activation , Complement C4/analysis , Diabetes Mellitus, Type 1/immunology , Adolescent , Adult , Autoantibodies/analysis , Biomarkers/analysis , Cadaver , Child , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/immunology , Female , Humans , Islets of Langerhans/immunology , Major Histocompatibility Complex/genetics , Male , Young Adult
8.
Am J Physiol Renal Physiol ; 304(7): F972-81, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23324176

ABSTRACT

The ammonia transporter family member, Rh B Glycoprotein (RhBG/Rhbg), is essential for ammonia transport by the rodent kidney, but in the human kidney mRNA but not protein expression has been reported. Because ammonia transport is fundamental for acid-base homeostasis, the current study addressed RhBG expression in the human kidney. Two distinct RhBG mRNA sequences have been reported, with different numbers of consecutive cytosines at nt1265 and thus encoding different carboxy-tails. Sequencing the region of difference in both human kidney and liver mRNA showed eight sequential cytosines, not seven as in some reports. Knowing the correct mRNA sequence for RhBG, we then assessed RhBG protein expression using antibodies against the correct amino acid sequence. Immunoblot analysis demonstrated RhBG protein expression in human kidney and immunohistochemistry identified basolateral RhBG in connecting segment (CNT) and the cortical and outer medullary collecting ducts. Colocalization of RhBG with multiple cell-specific markers demonstrated that that CNT cells and collecting duct type A intercalated cells express high levels of RhBG, and type B intercalated cells and principal cells do not express detectable RhBG. Thus, these studies identify the correct mRNA and thus protein sequence for human RhBG and show that the human kidney expresses basolateral RhBG protein in CNT, type A intercalated cells, and non-A, non-B cells. We conclude that RhBG can mediate an important role in human renal ammonia transport.


Subject(s)
Glycoproteins/biosynthesis , Kidney Tubules, Collecting/metabolism , Membrane Transport Proteins/biosynthesis , Amino Acid Sequence , Ammonia/metabolism , Animals , Base Sequence , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Kidney/metabolism , Liver/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/immunology , Mice , Mice, Knockout , Molecular Sequence Data , RNA, Messenger/analysis , RNA, Messenger/metabolism , Sequence Alignment
9.
J Physiol ; 591(5): 1313-24, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23266936

ABSTRACT

Exercise-induced vascular endothelial adaptations in the kidney are not well understood. Therefore, we investigated the impact of voluntary wheel running (VWR) on the abundance of endothelial nitric oxide synthase (eNOS) and extracellular superoxide dismutase (EC SOD), in kidney and lung, and other SOD isoforms and total antioxidant capacity (TAC), in kidney. We also determined whether VWR influences susceptibility to acute kidney injury (AKI). Male Sprague-Dawley and Fisher 344 rats, VWR or sedentary for 12 weeks, were subjected to AKI (uninephrectomy (UNX) and 35 min of left kidney ischaemia-24 h reperfusion, IR). We measured glomerular filtration rate (GFR) and renal plasma flow (RPF), and analysed renal structural injury. Running was comparable between strains and VWR reduced body weight. In Sprague-Dawley rats, VWR reduced eNOS and EC SOD, but increased Mn SOD in kidney. Similar changes were seen after 6 weeks of VWR in Sprague-Dawley rats. In Fisher 344 rats, VWR increased eNOS, all SOD isoforms and TAC in kidney. Both strains increased eNOS and EC SOD in lung with VWR. Compared to UNX alone, UNX-IR injury markedly reduced renal function for both strains; however, in the Sprague-Dawley rats, VWR exacerbated falls in GFR and RPF due to UNX-IR, whereas in the Fisher 344 rats, GFR was unaffected by VWR. Some indices of renal structural injury due to UNX-IR tended to be worse in SD vs. F344. Our study demonstrates that genetic background influences the effect of exercise on kidney eNOS and EC SOD, which in turn influence the susceptibility to AKI.


Subject(s)
Acute Kidney Injury/etiology , Kidney/metabolism , Physical Exertion , Reperfusion Injury/etiology , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Antioxidants/metabolism , Disease Models, Animal , Genotype , Glomerular Filtration Rate , Kidney/blood supply , Kidney/pathology , Kidney/physiopathology , Lung/metabolism , Male , Nitric Oxide Synthase Type III/metabolism , Phenotype , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Renal Plasma Flow , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Running , Species Specificity , Superoxide Dismutase/metabolism , Time Factors , Volition
10.
J Immunol ; 189(6): 2931-40, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22896639

ABSTRACT

The lupus-prone NZM2410 mice present an expanded B1a cell population that we have mapped to the Sle2c1 lupus susceptibility locus. The expression of Cdkn2c, a gene encoding for cyclin-dependent kinase inhibitor p18(Ink4c) and located within Sle2c1, is significantly lower in B6.Sle2c1 B cells than in B6 B cells. To test the hypothesis that the B1a cell expansion in B6.Sle2c1 mice was due to a defective p18 expression, we analyzed the B1a cell phenotypes of p18-deficient C57BL/6 mice. We found a dose-dependent negative correlation between the number of B1a cells and p18 expression in B cells, with p18-deficient mice showing an early expansion of the peritoneal B1a cell pool. p18 deficiency enhanced the homeostatic expansion of B1a cells but not of splenic conventional B cells, and the elevated number of B6.Sle2c1 B1a cells was normalized by cyclin D2 deficiency. These data demonstrated that p18 is a key regulator of the size of the B1a cell pool. B6.p18(-/-) mice produced significant amounts of anti-DNA IgM and IgG, indicating that p18 deficiency contributes to humoral autoimmunity. Finally, we have shown that Sle2c1 increases lpr-associated lymphadenopathy and T cell-mediated pathology. B6.p18(-/-).lpr mice showed a greater lymphadenopathy than B6.Sle2c1.lpr mice, but their renal pathology was intermediate between that of B6.lpr and B6.Sle2c1.lpr mice. This indicated that p18-deficiency synergizes, at least partially, with lpr-mediated pathology. These results show that Cdkn2c contributes to lupus susceptibility by regulating the size of the B1a cell compartment and hence their contribution to autoimmunity.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cell Differentiation/immunology , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinases/antagonists & inhibitors , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Animals , B-Lymphocyte Subsets/enzymology , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/physiology , Disease Models, Animal , Immunophenotyping , Lupus Erythematosus, Systemic/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Inbred NZB , Mice, Knockout , Mice, Transgenic
11.
Lab Invest ; 91(10): 1540-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21808234

ABSTRACT

Diffuse alveolar hemorrhage is an uncommon, yet often fatal, complication of systemic lupus erythematosus (SLE). Advances in the treatment of alveolar hemorrhage have been hampered because of the heterogeneity of clinical findings and the lack of suitable animal models. A single intraperitoneal injection of pristane induces a lupus-like syndrome characterized by lupus-related autoantibodies and glomerulonephritis in non-autoimmune-prone strains of mice. In addition, C57BL/6 (B6) mice frequently develop alveolar hemorrhage within a few weeks of pristane injection. Immunopathogenesis of pristane-induced alveolar hemorrhage was investigated in the present study. Early (2-4 weeks after injection) mortality due to hemorrhage was unique to C57BL/6 and C57BL/10 strains of mice. Recruitment of the macrophages and neutrophils preceded the hemorrhage by several days, and hemorrhage started 3-7 days after pristane injection in some mice, peaked at 2 weeks (84% in B6) and then resolved by 4 weeks in a majority of mice. Alveolar hemorrhage was independent of MyD88 (myeloid differentiation factor 88), or TLR7 pathways, in contrast to autoantibody production and glomerulonephritis, and was also independent of FcγR or Fas. Rag1(-/-) mice had a reduced prevalence of alveolar hemorrhage compared with B6 (P=0.01) congenics. However, T-cell receptor-deficient mice developed alveolar hemorrhage at a rate comparable to wild-type controls, whereas B6 Igµ(-/-) mice surprisingly had a strikingly reduced prevalence (7% vs 84% in B6, P<0.0001). Reconstitution of B6 Igµ(-/-) mice with wild-type B cells increased the prevalence to 50% (P=0.028). Pristane-induced alveolar hemorrhage is a useful model to study the pathogenesis and develop new therapy for this underappreciated and often life-threatening complication of SLE.


Subject(s)
B-Lymphocytes , Hemorrhage/chemically induced , Lung Diseases/chemically induced , Pulmonary Alveoli , Terpenes , Animals , B-Lymphocytes/pathology , Cell Line , Hemorrhage/pathology , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Lung/drug effects , Lung/pathology , Lung Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Species Specificity , Substrate Specificity
12.
J Immunol ; 186(12): 6673-82, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21543644

ABSTRACT

Sle2c1 is an NZM2410- and NZB-derived lupus susceptibility locus that induces an expansion of the B1a cell compartment. B1a cells have a repertoire enriched for autoreactivity, and an expansion of this B cell subset occurs in several mouse models of lupus. A combination of genetic mapping and candidate gene analysis presents Cdkn2c, a gene encoding for cyclin-dependent kinase inhibitor p18(INK4c) (p18), as the top candidate gene for inducing the Slec2c1-associated expansion of B1a cells. A novel single nucleotide polymorphism in the NZB allele of the Cdkn2c promoter is associated with a significantly reduced Cdkn2c expression in the splenic B cells and peritoneal cavity B1a cells from Sle2c1-carrying mice, which leads to a defective G1 cell cycle arrest in splenic B cells and increased proliferation of peritoneal cavity B1a cells. As the cell cycle is differentially regulated in B1a and B2 cells, these results suggest that Cdkn2c plays a critical role in B1a cell self-renewal and that its impaired expression leads to an accumulation of these cells with high autoreactive potential.


Subject(s)
B-Lymphocytes/pathology , Cyclin-Dependent Kinase Inhibitor p18/physiology , Genetic Predisposition to Disease/genetics , Homeostasis , Lupus Erythematosus, Systemic/pathology , Animals , Autoimmunity/genetics , B-Lymphocyte Subsets/pathology , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Cycle , Cell Proliferation , Chromosome Mapping , Cyclin-Dependent Kinase Inhibitor p18/genetics , Disease Models, Animal , Genetic Loci/genetics , Lupus Erythematosus, Systemic/genetics , Lymphocyte Count , Mice , Polymorphism, Single Nucleotide
13.
Arthritis Rheum ; 63(3): 764-74, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21360506

ABSTRACT

OBJECTIVE: Sle2 is a lupus susceptibility locus that has been linked to glomerulonephritis in the NZM2410 mouse. By itself, Sle2 does not induce any autoimmune pathology but results in the accumulation of B-1a cells. This study was designed to assess the contribution of Sle2 to the pathogenesis of autoimmunity. METHODS: Sle2 or its subcongenic intervals (Sle2a, Sle2b, and Sle2c1) were bred to Fas-deficient B6.lpr mice. Lymphoid phenotypes, which were focused on T cells, were assessed by flow cytometry, and histopathologic changes were compared between cohorts of B6.Sle2.lpr congenic mice and B6.lpr mice of ages up to 6 months. RESULTS: Sle2 synergized with lpr, resulting in a greatly accelerated lymphadenopathy that largely targeted T cells and mapped to the Sle2c1 locus. This locus has been identified as the main contributor to B-1a cell expansion. Further analyses showed that Sle2c1 expression skewed the differentiation and polarization of Fas-deficient T cells, with a reduction of the CD4+CD25+FoxP3+ regulatory T cell subset and an expansion of the Th17 cells. This was associated with a high number of T cell infiltrates that promoted severe nephritis and dermatitis in the B6.Sle2c1.lpr mice. CONCLUSION: These results show that Sle2c1 contributes to lupus pathogenesis by affecting T cell differentiation in combination with other susceptibility loci, such as lpr. The significance of the cosegregation of this phenotype and B-1a cell expansion in Sle2c1-expressing mice in relation to the pathogenesis of lupus is discussed.


Subject(s)
Cell Polarity/immunology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Th17 Cells/immunology , fas Receptor/genetics , Animals , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/pathology , Female , Genetic Predisposition to Disease/genetics , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Kidney/immunology , Kidney/pathology , Lupus Nephritis/pathology , Lymphatic Diseases/genetics , Lymphatic Diseases/immunology , Lymphatic Diseases/pathology , Male , Mice , Mice, Congenic , Mice, Inbred MRL lpr , Mice, Inbred NZB , Mice, Transgenic , Spleen/immunology , Spleen/pathology , Th17 Cells/pathology
14.
Mech Ageing Dev ; 132(1-2): 1-7, 2011.
Article in English | MEDLINE | ID: mdl-21087620

ABSTRACT

Age-dependent renal damage is influenced by genetic background and the Fisher344xBrown Norway (F344xBN) rat is resistant to glomerular injury. In vulnerable strains, a fall in renal nitric oxide synthase (NOS) contributes to age-dependent renal damage. Here, we investigated renal NOS in young (3 months) and old (30 months) male F344xBN to test the hypothesis that renal NOS is maintained in "protected" strains. We also examined if 6 months of renin-angiotensin system (RAS) blockade using angiotensin converting enzyme inhibition (ACEI) and angiotensin receptor blockade (ARB) provides further benefit in these "protected" old rats. Aging increased tubulointerstitial injury but glomerular sclerosis was minimal and NOS and superoxide dismutase abundance increased. There was no change in the NOS inhibitor, ADMA (asymmetric dimethylarginine) or its regulatory enzymes. RAS blockade with ARB protected against tubulointerstitial injury and increased nNOSα, but ACEI, which also increased nNOSα, had no protective effect on the tubulointerstitium. We conclude that the glomerular sclerosis-resistant aged male F344xBN rat maintains renal NOS, thus reinforcing our hypothesis that progressive glomerular injury is related to renal NOS deficiency. The tubulointerstitial injury seen with aging is reversed with 6 months of ARB but not ACEI and is not associated with renal NOS.


Subject(s)
Aging/metabolism , Aging/pathology , Kidney/enzymology , Kidney/pathology , Nitric Oxide Synthase/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Kidney/drug effects , Kidney/physiopathology , Male , Models, Animal , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase Type I , Oxidative Stress , Rats , Rats, Inbred BN , Rats, Inbred F344 , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Species Specificity
15.
Nat Rev Nephrol ; 7(1): 36-44, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21045790

ABSTRACT

The mechanisms that drive the development of diabetic nephropathy remain undetermined. Only 30-40% of patients with diabetes mellitus develop overt nephropathy, which suggests that other contributing factors besides the diabetic state are required for the progression of diabetic nephropathy. Endothelial dysfunction is associated with human diabetic nephropathy and retinopathy, and advanced diabetic glomerulopathy often exhibits thrombotic microangiopathy, including glomerular capillary microaneurysms and mesangiolysis, which are typical manifestations of endothelial dysfunction in the glomerulus. Likewise, diabetic mice with severe endothelial dysfunction owing to deficiency of endothelial nitric oxide synthase develop progressive nephropathy and retinopathy similar to the advanced lesions observed in humans with diabetes mellitus. Additionally, inhibitors of the renin-angiotensin system fail to be renoprotective in some individuals with diabetic nephropathy (due in part to aldosterone breakthrough) and in some mouse models of the disease. In this Review, we discuss the clinical and experimental evidence that supports a role for endothelial nitric oxide deficiency and subsequent endothelial dysfunction in the progression of diabetic nephropathy and retinopathy. If endothelial dysfunction is the key factor required for diabetic nephropathy, then agents that improve endothelial function or raise intraglomerular nitric oxide level could be beneficial in the treatment of diabetic nephropathy.


Subject(s)
Diabetic Angiopathies/physiopathology , Diabetic Nephropathies/physiopathology , Endothelium, Vascular/physiopathology , Animals , Diabetic Angiopathies/epidemiology , Diabetic Angiopathies/therapy , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/therapy , Disease Models, Animal , Humans , Mice
16.
Am J Nephrol ; 32(6): 575-80, 2010.
Article in English | MEDLINE | ID: mdl-21071934

ABSTRACT

BACKGROUND/AIMS: the C57Bl6 mouse is resistant to chronic kidney disease (CKD) induced by reduction of renal mass (RRM). Nitric oxide (NO) deficiency exacerbates CKD progression so this study investigated whether combination of RRM and NO deficiency would render the C57Bl6 mouse vulnerable to CKD. METHODS: we used wild-type (WT) mice with RRM, chronic NO synthase (NOS) inhibition and a combination. Also, endothelial NOS (eNOS) knockout (KO) C57Bl6 mice were studied with and without RRM. Primary endpoints were albuminuria and structural damage. RESULTS: both nonselective (+L-NAME) and neuronal NOS 'selective' (+7NI) NOS inhibition greatly exacerbated the albuminuria and structural damage seen with RRM in the WT mice; NOS inhibition alone had little effect. The eNOS KO mice showed marked structural damage and significant albuminuria in the shams and RRM produced minimal exacerbation of structural damage although the albuminuria was massively amplified. CONCLUSION: these studies demonstrate that the C57Bl6 mouse is rendered vulnerable to RRM-induced CKD when concomitant NO deficiency is produced. This observation supports previous work in CKD-resistant rats and suggests that NO deficiency is required for progression of CKD.


Subject(s)
Kidney Failure, Chronic/pathology , Kidney/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide/deficiency , Albuminuria , Animals , Disease Progression , Epithelial Cells/metabolism , Kidney/metabolism , Kidney Failure, Chronic/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism
17.
Am J Pathol ; 176(5): 2198-208, 2010 May.
Article in English | MEDLINE | ID: mdl-20363914

ABSTRACT

Endothelial dysfunction is critical in the decline of renal function with. By using endothelial nitric oxide synthase knockout (eNOSKO) mice, we tested the hypothesis that a lack of endothelial nitric oxide synthase accelerates renal injury in the aging kidney. In contrast to control mice and young eNOSKO mice, aging eNOSKO mice showed greater renal injury and in particular developed a thrombotic microangiopathy, with mesangiolysis, endothelial swelling, endothelial cell loss, double-contour appearance of glomerular basement membrane (GBM), and thrombus formation. Thrombi, which were composed of fibrin, platelets, and von Willebrand factor (vWF), were identified predominantly in glomerular capillaries and rarely in arterioles, but not in larger vessels. In the tubulointerstitium, tubular degeneration and macrophage infiltration were also prominent in aging eNOSKO mice. Intraluminal vWF deposition was accompanied with thrombus formation, whereas mesangial deposition of vWF was associated with mesangial matrix expansion. Furthermore, the mesangial vWF deposition was detectable in young eNOSKO mice in which severe glomerular injury had not yet developed. Finally, a higher level of serum P-selectin in eNOSKO mice was consistent with the vWF behavior and suggested exocytosis of the Weibel-Palade body by the endothelium. In conclusion, a lack of endothelial nitric oxide synthase resulted in the development of glomerular thrombotic microangiopathy. A lack of nitric oxide likely contributed to the release of vWF, leading to thrombus formation in this model.


Subject(s)
Aging , Endothelium, Vascular/metabolism , Gene Expression Regulation, Enzymologic , Kidney/metabolism , Nitric Oxide Synthase Type III/metabolism , Thrombotic Microangiopathies/metabolism , von Willebrand Factor/metabolism , Animals , Exocytosis , Male , Mice , Mice, Knockout , P-Selectin/blood , Thrombosis , Weibel-Palade Bodies/pathology
18.
Am J Physiol Renal Physiol ; 298(3): F609-16, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20015944

ABSTRACT

VEGF is recognized as a major mediator in the development of diabetic nephropathy. Soluble Flt-1 (sFlt-1) is the endogenous inhibitor of VEGF, and recently genetic overexpression of sFlt-1 in the podocyte was shown to be protective in murine diabetic nephropathy. In this study, we performed a translational study to determine whether an intramuscular gene transfer of sFlt-1 can prevent the progression of renal disease in diabetic db/db mice. Adeno-associated virus-1 (AAV1) encoding human sFlt-1 in two different doses was intramuscularly administrated in db/db and wild-type mice. The sFlt-1-AAV1 treatment significantly increased serum sFlt-1 level at 4 and 8 wk. A dose that was developed in this study caused minimal abnormalities in normal mice but reduced albuminuria in diabetic db/db mice. In renal histology, sFlt-1 treatment at this dose had minimal effects on mesangial expansion in diabetic mice, whereas podocyte injury was significantly improved, at 8 wk. Unfortunately, tubulointerstitial injury was markedly exacerbated by sFlt-1 treatment in association with a reduction in endogenous VEGF expression and peritubular capillary loss. In conclusion, gene therapy with sFlt-1-AAV1 protects podocytes but accelerates tubulointerstitial injury in diabetic db/db mice. These data suggest systemic overexpression of sFlt-1 will not likely be useful for treating diabetic nephropathy.


Subject(s)
Albuminuria/therapy , Diabetic Nephropathies/therapy , Genetic Therapy , Kidney/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Albuminuria/genetics , Albuminuria/metabolism , Albuminuria/pathology , Animals , Capillaries/metabolism , Capillaries/pathology , Collagen/metabolism , Dependovirus/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Models, Animal , Genetic Therapy/adverse effects , Genetic Vectors , Humans , Injections, Intramuscular , Kidney/blood supply , Kidney/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Podocytes/metabolism , Podocytes/pathology , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/urine , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/genetics
19.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R740-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20018820

ABSTRACT

Recent studies have shown that asymmetric dimethylarginine (ADMA), a nitric oxide synthase inhibitor, is increased in hypertension and chronic kidney disease. However, little is known about the effects of hypertension per se on ADMA metabolism. The purpose of this study was to test the hypothesis that ANG II-induced hypertension, in the absence of renal injury, is associated with increased oxidative stress and plasma and renal cortex ADMA levels in rats. Male Sprague-Dawley rats were treated with ANG II at 200 ng.kg(-1).min(-1) sc (by minipump) for 1 or 3 wk or at 400 ng.kg(-1).min(-1) for 6 wk. Mean arterial pressure was increased after 3 and 6 wk of ANG II; however, renal injury (proteinuria, glomerular sclerosis, and interstitial fibrosis) was only evident after 6 wk of treatment. Plasma thiobarbituric acid reactive substances concentration and renal cortex p22(phox) protein abundance were increased early (1 and 3 wk), but urinary excretion of isoprostane and H(2)O(2) was only increased after 6 wk of ANG II. An increased in plasma ADMA after 6 wk of ANG II was associated with increased lung protein arginine methyltransferase-1 abundance and decreased renal cortex dimethylarginine dimethylaminohydrolase activity. No changes in renal cortex ADMA were observed. ANG II hypertension in the absence of renal injury is not associated with increased ADMA; however, when the severity and duration of the treatment were increased, plasma ADMA increased. These data suggest that elevated blood pressure alone, for up to 3 wk, in the absence of renal injury does not play an important role in the regulation of ADMA. However, the presence of renal injury and sustained hypertension for 6 wk increases ADMA levels and contributes to nitric oxide deficiency and cardiovascular disease.


Subject(s)
Arginine/analogs & derivatives , Hypertension, Renal/metabolism , Kidney Cortex/metabolism , Oxidative Stress/physiology , Amidohydrolases/metabolism , Angiotensin II/pharmacology , Animals , Arginine/blood , Arginine/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Fibrosis , Hypertension, Renal/chemically induced , Hypertension, Renal/pathology , Kidney Cortex/pathology , Male , NADPH Oxidases/metabolism , Nitrates/urine , Nitrites/urine , Protein-Arginine N-Methyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism , Vasoconstrictor Agents/pharmacology
20.
Am J Pathol ; 174(4): 1221-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19246639

ABSTRACT

Recently, we and others reported that diabetic endothelial nitric oxide synthase knockout (eNOSKO) mice develop advanced glomerular lesions that include mesangiolysis and nodular lesions. Interestingly, insulin treatment lowered blood pressure and prevented renal lesions, raising the question as to whether these beneficial effects of insulin were due to its ability to lower either high glucose levels or high blood pressure. We, therefore, examined the effect of lowering blood pressure using hydralazine in this diabetic eNOSKO mouse model. Hydralazine treatment significantly blocked the development of mesangiolysis and microaneurysms, whereas tubulointerstitial injury was not prevented in these mice. Additionally, hydralazine did not reduce expression levels of either tubulointerstitial thrombospondin-1 or transforming growth factor-beta despite controlling blood pressure. On the other hand, the critical role of high glucose levels on the development of tubulointerstitial injury was suggested by the observation that serum glucose levels were correlated with tubulointerstitial injury, as well as with the expression levels of both transforming growth factor-beta and thrombospondin-1. Importantly, controlling blood glucose with insulin completely blocked tubulointerstitial injury in diabetic eNOSKO mice. These data suggest that glomerular injury is dependent on systemic blood pressure, whereas hyperglycemia may have a more important role in tubulointerstitial injury, possibly due to the stimulation of the thrombospondin-1-transforming growth factor-beta pathway in diabetic eNOSKO mice. This study could provide insights into the pathogenesis of advanced diabetic nephropathy in the presence of endothelial dysfunction.


Subject(s)
Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Nitric Oxide Synthase Type III/genetics , Animals , Blotting, Western , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Glomerular Mesangium/drug effects , Glomerular Mesangium/pathology , Hydralazine/therapeutic use , Male , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/biosynthesis , Thrombospondin 1/drug effects , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...