Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 371, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191531

ABSTRACT

Aurora Kinase A (AURKA) promotes cell proliferation and is overexpressed in different types of polycystic kidney disease (PKD). To understand AURKA's role in regulating renal cyst development we conditionally deleted the gene in mouse models of Autosomal Dominant PKD (ADPKD) and Joubert Syndrome, caused by Polycystin 1 (Pkd1) and Inositol polyphosphate-5-phosphatase E (Inpp5e) mutations respectively. We show that while Aurka is dispensable for collecting duct development and homeostasis, its deletion prevents cyst formation in both disease models. Cross-comparison of transcriptional changes implicated AKT signaling in cyst prevention and we show that (i) AURKA and AKT physically interact, (ii) AURKA regulates AKT activity in a kinase-independent manner and (iii) inhibition of AKT can reduce disease severity. AKT activation also regulates Aurka expression, creating a feed-forward loop driving renal cystogenesis. We find that the AURKA kinase inhibitor Alisertib stabilises the AURKA protein, agonizing its cystogenic functions. These studies identify AURKA as a master regulator of renal cyst development in different types of PKD, functioning in-part via AKT.


Subject(s)
Aurora Kinase A , Cysts , Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Animals , Mice , Aurora Kinase A/genetics , Phosphoric Monoester Hydrolases , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/prevention & control , Proto-Oncogene Proteins c-akt/genetics
2.
Kidney Int ; 105(4): 731-743, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38158181

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a leading cause of kidney failure and is associated with substantial morbidity and mortality. Interstitial inflammation is attributed to the action of infiltrating macrophages and is a feature thought to aggravate disease progression. Here, we investigated the therapeutic potential of the anti-inflammatory IL37b cytokine as a treatment for ADPKD using genetic mouse models, demonstrating that transgenic expression of human IL37b reduced collecting duct cyst burden in both early and adult-onset ADPKD rodent models. Moreover, injection of recombinant human IL37b could also reduce cyst burden in early onset ADPKD mice, an observation not associated with increased macrophage number at early stages of cyst formation. Interestingly, transgenic IL37b expression also did not alter macrophage numbers in advanced disease. Whole kidney RNA-seq highlighted an IL37b-mediated upregulation of the interferon signaling pathway and single-cell RNA-seq established that these changes originate at least partly from kidney resident macrophages. We further found that blocking type I interferon signaling in mice expressing IL37b resulted in increased cyst number, confirming this as an important pathway by which IL37b exerts its beneficial effects. Thus, our studies show that IL37b promotes interferon signaling in kidney resident macrophages which suppresses cyst initiation, identifying this protein as a potential therapy for ADPKD.


Subject(s)
Cysts , Polycystic Kidney, Autosomal Dominant , Mice , Humans , Animals , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/genetics , Inflammation/genetics , Inflammation/complications , Kidney/metabolism , Cysts/complications , Interleukins , Interferons
3.
bioRxiv ; 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38106143

ABSTRACT

Low nephron number correlates with the development of hypertension and chronic kidney disease later in life. While intrauterine growth restriction caused by maternal low protein diet (LPD) is thought to be a significant cause of reduced nephron endowment in impoverished communities, its influence on the cellular and molecular processes which drive nephron formation are poorly understood. We conducted a comprehensive characterization of the impact of LPD on kidney development using tomographic and confocal imaging to quantify changes in branching morphogenesis and the cellular and morphological features of nephrogenic niches across development. These analyses were paired with single-cell RNA sequencing to dissect the transcriptional changes that LPD imposes during renal development. Differences in the expression of genes involved in metabolism were identified in most cell types we analyzed, yielding imbalances and shifts in cellular energy production. We further demonstrate that LPD impedes branching morphogenesis and significantly reduces the number of pretubular aggregates - the initial precursors to nephron formation. The most striking observation was that LPD changes the developmental trajectory of nephron progenitor cells, driving the formation of a partially committed cell population which likely reflects a failure of cells to commit to nephron formation and which ultimately reduces endowment. This unique profile of a fetal programming defect demonstrates that low nephron endowment arises from the pleiotropic impact of changes in branching morphogenesis and nephron progenitor cell commitment, the latter of which highlights a critical role for nutrition in regulating the cell fate decisions underpinning nephron endowment. Significance Statement: While a mother's diet and behavior can negatively impact the number of nephrons in the kidneys of her offspring, the root cellular and molecular drivers of these deficits have not been rigorously explored. In this study we use advanced imaging and gene expression analysis in mouse models to define how a maternal low protein diet, analogous to that of impoverished communities, results in reduced nephron endowment. We find that low protein diet has pleiotropic effects on metabolism and the normal programs of gene expression. These profoundly impact the process of branching morphogenesis necessary to establish niches for nephron generation and change cell behaviors which regulate how and when nephron progenitor cells commit to differentiation.

4.
Cell Rep ; 42(5): 112322, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37105170

ABSTRACT

Crosstalk between cardiac cells is critical for heart performance. Here we show that vascular cells within human cardiac organoids (hCOs) enhance their maturation, force of contraction, and utility in disease modeling. Herein we optimize our protocol to generate vascular populations in addition to epicardial, fibroblast, and cardiomyocyte cells that self-organize into in-vivo-like structures in hCOs. We identify mechanisms of communication between endothelial cells, pericytes, fibroblasts, and cardiomyocytes that ultimately contribute to cardiac organoid maturation. In particular, (1) endothelial-derived LAMA5 regulates expression of mature sarcomeric proteins and contractility, and (2) paracrine platelet-derived growth factor receptor ß (PDGFRß) signaling from vascular cells upregulates matrix deposition to augment hCO contractile force. Finally, we demonstrate that vascular cells determine the magnitude of diastolic dysfunction caused by inflammatory factors and identify a paracrine role of endothelin driving dysfunction. Together this study highlights the importance and role of vascular cells in organoid models.


Subject(s)
Endothelial Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Pericytes/metabolism , Signal Transduction , Organoids/metabolism
5.
Cell Rep Med ; 1(8): 100129, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33294854

ABSTRACT

Mutations in the lipid transport protein ABCA12 cause the life-threatening skin condition harlequin ichthyosis (HI), which is characterized by the loss of skin barrier function, inflammation, and dehydration. Inflammatory responses in HI increase disease severity by impairing keratinocyte differentiation, suggesting amelioration of this phenotype as a possible therapy for the condition. Existing treatments for HI are based around the use of retinoids, but their value in treating patients during the neonatal period has been questioned relative to other improved management regimens, and their long-term use is associated with side effects. We have developed a conditional mouse model to demonstrate that topical application of the aminosalicylic acid derivatives 5ASA or 4ASA considerably improves HI keratinocyte differentiation without the undesirable side effects of the retinoid acitretin and salicylic acid (aspirin). Analysis of changes in gene expression shows that 4ASA in particular elicits compensatory upregulation of a large family of barrier function-related genes, many of which are associated with other ichthyoses, identifying this compound as a lead candidate for developing topical treatments for HI.


Subject(s)
Aminosalicylic Acid/pharmacology , Cell Differentiation/drug effects , Ichthyosis, Lamellar/drug therapy , Keratinocytes/drug effects , ATP-Binding Cassette Transporters/metabolism , Acitretin/pharmacology , Animals , Disease Models, Animal , Epidermis/drug effects , Epidermis/metabolism , Gene Expression/drug effects , Ichthyosis, Lamellar/metabolism , Keratinocytes/metabolism , Mice , Mice, Knockout , Mutation/drug effects , Phenotype , Salicylic Acid/pharmacology , Skin/drug effects , Skin/metabolism , Up-Regulation/drug effects
6.
Development ; 147(21)2020 06 22.
Article in English | MEDLINE | ID: mdl-32439764

ABSTRACT

Laminin alpha 5 (LAMA5) is a member of a large family of proteins that trimerise and then polymerise to form a central component of all basement membranes. Consequently, the protein plays an instrumental role in shaping the normal development of the kidney, skin, neural tube, lung and limb, and many other organs and tissues. Pathogenic mutations in some laminins have been shown to cause a range of largely syndromic conditions affecting the competency of the basement membranes to which they contribute. We report the identification of a mutation in the polymerisation domain of LAMA5 in a patient with a complex syndromic disease characterised by defects in kidney, craniofacial and limb development, and by a range of other congenital defects. Using CRISPR-generated mouse models and biochemical assays, we demonstrate the pathogenicity of this variant, showing that the change results in a failure of the polymerisation of α/ß/γ laminin trimers. Comparing these in vivo phenotypes with those apparent upon gene deletion in mice provides insights into the specific functional importance of laminin polymerisation during development and tissue homeostasis.


Subject(s)
Developmental Disabilities/genetics , Fetal Development , Laminin/genetics , Mutation/genetics , Polymerization , Amino Acid Sequence , Animals , Animals, Newborn , Child, Preschool , Developmental Disabilities/pathology , Fetus/embryology , Humans , Hydronephrosis/pathology , Infant, Newborn , Kidney/abnormalities , Kidney/embryology , Kidney/pathology , Laminin/chemistry , Lung/abnormalities , Lung/embryology , Lung/pathology , Male , Mice , Protein Domains , Syndrome
7.
EMBO Rep ; 21(3): e48692, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32072744

ABSTRACT

Dysregulation of lipid homeostasis is intimately associated with defects in insulin secretion, a key feature of type 2 diabetes. Here, we explore the role of the putative lipid transporter ABCA12 in regulating insulin secretion from ß-cells. Mice with ß-cell-specific deletion of Abca12 display impaired glucose-stimulated insulin secretion and eventual islet inflammation and ß-cell death. ABCA12's action in the pancreas is independent of changes in the abundance of two other cholesterol transporters, ABCA1 and ABCG1, or of changes in cellular cholesterol or ceramide content. Instead, loss of ABCA12 results in defects in the genesis and fusion of insulin secretory granules and increases in the abundance of lipid rafts at the cell membrane. These changes are associated with dysregulation of the small GTPase CDC42 and with decreased actin polymerisation. Our findings establish a new, pleiotropic role for ABCA12 in regulating pancreatic lipid homeostasis and insulin secretion.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice
8.
Cell Rep ; 24(12): 3285-3295.e4, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30232009

ABSTRACT

Apoptotic cell death removes unwanted cells and is regulated by interactions between pro-survival and pro-apoptotic members of the BCL-2 protein family. The regulation of apoptosis is thought to be crucial for normal embryonic development. Accordingly, complete loss of pro-survival MCL-1 or BCL-XL (BCL2L1) causes embryonic lethality. However, it is not known whether minor reductions in pro-survival proteins could cause developmental abnormalities. We explored the rate-limiting roles of MCL-1 and BCL-XL in development and show that combined loss of single alleles of Mcl-1 and Bcl-x causes neonatal lethality. Mcl-1+/-;Bcl-x+/- mice display craniofacial anomalies, but additional loss of a single allele of pro-apoptotic Bim (Bcl2l11) restores normal development. These findings demonstrate that the control of cell survival during embryogenesis is finely balanced and suggest that some human craniofacial defects, for which causes are currently unknown, may be due to subtle imbalances between pro-survival and pro-apoptotic BCL-2 family members.


Subject(s)
Bcl-2-Like Protein 11/genetics , Craniofacial Abnormalities/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , bcl-X Protein/genetics , Animals , Apoptosis , Bcl-2-Like Protein 11/metabolism , Cells, Cultured , Female , Heterozygote , Male , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , bcl-X Protein/metabolism
9.
Elife ; 72018 07 31.
Article in English | MEDLINE | ID: mdl-30063208

ABSTRACT

Branching morphogenesis of the ureteric bud is integral to kidney development; establishing the collecting ducts of the adult organ and driving organ expansion via peripheral interactions with nephron progenitor cells. A recent study suggested that termination of tip branching within the developing kidney involved stochastic exhaustion in response to nephron formation, with such a termination event representing a unifying developmental process evident in many organs. To examine this possibility, we have profiled the impact of nephron formation and maturation on elaboration of the ureteric bud during mouse kidney development. We find a distinct absence of random branch termination events within the kidney or evidence that nephrogenesis impacts the branching program or cell proliferation in either tip or progenitor cell niches. Instead, organogenesis proceeds in a manner indifferent to the development of these structures. Hence, stochastic cessation of branching is not a unifying developmental feature in all branching organs.


Subject(s)
Nephrons/embryology , Organogenesis , Animals , Cell Proliferation , Embryo, Mammalian/metabolism , Green Fluorescent Proteins/metabolism , Mice , Nephrons/cytology , Ureter/embryology
10.
Hum Mol Genet ; 24(2): 436-49, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25209981

ABSTRACT

Harlequin ichthyosis (HI) is a severe skin disease which leads to neonatal death in ∼50% of cases. It is the result of mutations in ABCA12, a protein that transports lipids required to establish the protective skin barrier needed after birth. To better understand the life-threatening newborn HI phenotype, we analysed the developing epidermis for consequences of lipid dysregulation in mouse models. We observed a pro-inflammatory signature which was characterized by chemokine upregulation in embryonic skin which is distinct from that seen in other types of ichthyosis. Inflammation also persisted in grafted HI skin. To examine the contribution of inflammation to disease development, we overexpressed interleukin-37b to globally suppress fetal inflammation, observing considerable improvements in keratinocyte differentiation. These studies highlight inflammation as an unexpected contributor to HI disease development in utero, and suggest that inhibiting inflammation may reduce disease severity.


Subject(s)
Ichthyosis, Lamellar/embryology , Ichthyosis, Lamellar/immunology , Animals , Cell Differentiation , Chemokines/genetics , Chemokines/immunology , Disease Models, Animal , Epidermis/embryology , Epidermis/immunology , Female , Humans , Ichthyosis, Lamellar/genetics , Ichthyosis, Lamellar/physiopathology , Interleukin-1/genetics , Interleukin-1/immunology , Keratinocytes/cytology , Male , Mice , Mice, Knockout , Phenotype , Skin/embryology , Skin/immunology
11.
PLoS Genet ; 10(10): e1004705, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25340873

ABSTRACT

The skin is a highly regenerative organ which plays critical roles in protecting the body and sensing its environment. Consequently, morbidity and mortality associated with skin defects represent a significant health issue. To identify genes important in skin development and homeostasis, we have applied a high throughput, multi-parameter phenotype screen to the conditional targeted mutant mice generated by the Wellcome Trust Sanger Institute's Mouse Genetics Project (Sanger-MGP). A total of 562 different mouse lines were subjected to a variety of tests assessing cutaneous expression, macroscopic clinical disease, histological change, hair follicle cycling, and aberrant marker expression. Cutaneous lesions were associated with mutations in 23 different genes. Many of these were not previously associated with skin disease in the organ (Mysm1, Vangl1, Trpc4ap, Nom1, Sparc, Farp2, and Prkab1), while others were ascribed new cutaneous functions on the basis of the screening approach (Krt76, Lrig1, Myo5a, Nsun2, and Nf1). The integration of these skin specific screening protocols into the Sanger-MGP primary phenotyping pipelines marks the largest reported reverse genetic screen undertaken in any organ and defines approaches to maximise the productivity of future projects of this nature, while flagging genes for further characterisation.


Subject(s)
Mutation/genetics , Phenotype , Skin Physiological Phenomena/genetics , Animals , Embryonic Stem Cells , Hair Follicle/metabolism , Hair Follicle/physiology , Mice , Reverse Genetics
12.
Dis Model Mech ; 6(6): 1426-33, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24046351

ABSTRACT

Fras1-related extracellular matrix protein 1 (FREM1) is required for epidermal adhesion during embryogenesis, and mice lacking the gene develop fetal skin blisters and a range of other developmental defects. Mutations in members of the FRAS/FREM gene family cause diseases of the Fraser syndrome spectrum. Embryonic epidermal blistering is also observed in mice lacking PdgfC and its receptor, PDGFRα. In this article, we show that FREM1 binds to PDGFC and that this interaction regulates signalling downstream of PDGFRα. Fibroblasts from Frem1-mutant mice respond to PDGFC stimulation, but with a shorter duration and amplitude than do wild-type cells. Significantly, PDGFC-stimulated expression of the metalloproteinase inhibitor Timp1 is reduced in cells with Frem1 mutations, leading to reduced basement membrane collagen I deposition. These results show that the physical interaction of FREM1 with PDGFC can regulate remodelling of the extracellular matrix downstream of PDGFRα. We propose that loss of FREM1 function promotes epidermal blistering in Fraser syndrome as a consequence of reduced PDGFC activity, in addition to its stabilising role in the basement membrane.


Subject(s)
Extracellular Matrix Proteins/physiology , Extracellular Matrix/metabolism , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Signal Transduction/physiology , Animals , Extracellular Matrix Proteins/metabolism , Mice , NIH 3T3 Cells , Protein Binding
13.
Cell Metab ; 18(2): 225-38, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23931754

ABSTRACT

ABCA12 is involved in the transport of ceramides in skin, but it may play a wider role in lipid metabolism. We show that, in Abca12-deficient macrophages, cholesterol efflux failed to respond to activation with LXR agonists. Abca12 deficiency caused a reduction in the abundance of Abca1, Abcg1, and Lxrß. Overexpression of Lxrß reversed the effects. Mechanistically, Abca12 deficiency did not affect expression of genes involved in cholesterol metabolism. Instead, a physical association between Abca1, Abca12, and Lxrß proteins was established. Abca12 deficiency enhanced interaction between Abca1 and Lxrß and the degradation of Abca1. Overexpression of ABCA12 in HeLa-ABCA1 cells increased the abundance and stability of ABCA1. Abca12 deficiency caused an accumulation of cholesterol in macrophages and the formation of foam cells, impaired reverse cholesterol transport in vivo, and increased the development of atherosclerosis in irradiated Apoe(-/-) mice reconstituted with Apoe(-/-)Abca12(-/-) bone marrow. Thus, ABCA12 regulates the cellular cholesterol metabolism via an LXRß-dependent posttranscriptional mechanism.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , Macrophages/metabolism , Orphan Nuclear Receptors/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/deficiency , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Biological Transport/genetics , Cell Line , Foam Cells/metabolism , HeLa Cells , Humans , Lipid Metabolism/genetics , Lipoproteins/metabolism , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orphan Nuclear Receptors/biosynthesis
14.
PLoS One ; 8(3): e58243, 2013.
Article in English | MEDLINE | ID: mdl-23516451

ABSTRACT

There is strong evidence from human and animal models that exposure to maternal hyperglycemia during in utero development can detrimentally affect fetal kidney development. Notwithstanding this knowledge, the precise effects of diabetic pregnancy on the key processes of kidney development are unclear due to a paucity of studies and limitations in previously used methodologies. The purpose of the present study was to elucidate the effects of hyperglycemia on ureteric branching morphogenesis and nephrogenesis using unbiased techniques. Diabetes was induced in pregnant C57Bl/6J mice using multiple doses of streptozotocin (STZ) on embryonic days (E) 6.5-8.5. Branching morphogenesis was quantified ex vivo using Optical Projection Tomography, and nephrons were counted using unbiased stereology. Maternal hyperglycemia was recognised from E12.5. At E14.5, offspring of diabetic mice demonstrated fetal growth restriction and a marked deficit in ureteric tip number (control 283.7 ± 23.3 vs. STZ 153.2 ± 24.6, mean ± SEM, p<0.01) and ureteric tree length (control 33.1 ± 2.6 mm vs. STZ 17.6 ± 2.7 mm, p = 0.001) vs. controls. At E18.5, fetal growth restriction was still present in offspring of STZ dams and a deficit in nephron endowment was observed (control 1246.2 ± 64.9 vs. STZ 822.4 ± 74.0, p<0.001). Kidney malformations in the form of duplex ureter and hydroureter were a common observation (26%) in embryos of diabetic pregnancy compared with controls (0%). Maternal insulin treatment from E13.5 normalised maternal glycaemia but did not normalise fetal weight nor prevent the nephron deficit. The detrimental effect of hyperglycemia on ureteric branching morphogenesis and, in turn, nephron endowment in the growth-restricted fetus highlights the importance of glycemic control in early gestation and during the initial stages of renal development.


Subject(s)
Hyperglycemia/metabolism , Morphogenesis , Nephrons/embryology , Pregnancy in Diabetics/metabolism , Ureter/embryology , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Experimental , Female , Fetal Development/drug effects , Humans , Hyperglycemia/chemically induced , Insulin/metabolism , Insulin/pharmacology , Mice , Morphogenesis/drug effects , Nephrons/drug effects , Pregnancy , Pregnancy in Diabetics/chemically induced , Prenatal Exposure Delayed Effects , Streptozocin/adverse effects , Ureter/drug effects , Urinary Tract/abnormalities , Urinary Tract/embryology
15.
Nephrology (Carlton) ; 14(7): 650-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19796023

ABSTRACT

AIM: Statins are widely used for their cholesterol-lowering effects and for prevention of cardiovascular disease. Evidence indicates that these drugs also have immunomodulatory and other non-lipid lowering effects, with studies suggesting benefit in some animal models of immune (particularly T helper (Th)1)-mediated inflammatory disease and their corresponding human disease counterparts. We sought to evaluate the immunomodulatory effects and therapeutic potential of atorvastatin in experimental crescentic glomerulonephritis, a Th1-predominant animal model of glomerulonephritis. METHODS: Autologous phase, anti-glomerular basement membrane glomerulonephritis was induced in C57BL/6 mice by intravenous injection of sheep anti-mouse glomerular basement membrane globulin. Mice were administered atorvastatin (10 or 100 mg/kg) or control (phosphate-buffered saline) daily by oral gavage. Immune responses and renal injury were assessed after 21 days. RESULTS: Compared with control-treated mice, treatment with atorvastatin did not alter renal injury (serum creatinine, proteinuria, glomerular crescent formation) or glomerular leukocytic infiltration (CD4(+) T cells or macrophages). Atorvastatin resulted in a dose-related increase in circulating serum antibody to the disease-inducing antigen but no differences in antigen-stimulated splenocyte production of Th1/Th2 cytokines. At the higher dose, atorvastatin also led to a significant reduction in apoptosis of splenic CD4(+) T lymphocytes. CONCLUSION: This study demonstrates that statins modulate humoral responses and alter splenic CD4(+) T cell apoptosis. However, atorvastatin does not lead to significant changes in T helper cell polarization or renal injury in experimental crescentic glomerulonephritis.


Subject(s)
Glomerulonephritis/drug therapy , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunity, Humoral/drug effects , Kidney/drug effects , Pyrroles/pharmacology , Animals , Atorvastatin , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Drug , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Kidney/pathology , Male , Mice , Mice, Inbred C57BL
16.
Immunology ; 128(1): 114-22, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19689741

ABSTRACT

CD100 participates in adaptive immune responses and is important in neural cell migration. To determine the role of endogenous CD100 in severe glomerular inflammation, we induced experimental crescentic glomerulonephritis by planting a foreign antigen in glomeruli of sensitized normal and CD100-deficient (CD100(-/-)) mice. Fewer CD100(-/-) glomeruli exhibited crescent formation or severe histological changes. Antigen-specific immune responses were reduced in CD100(-/-) mice. There was less interferon (IFN)-gamma and interleukin (IL)-4 production by splenocytes and fewer activated T and B cells were present in lymph nodes of immunized CD100(-/-) mice. Serum antigen-specific immunoglobulin (IgG) levels were also decreased. Glomerular macrophage and CD4(+) cell infiltration, and IgG and C3 deposition were attenuated. Normal kidneys expressed mRNA for CD100 and plexin-B1 (the tissue receptor of CD100). Direct immunofluorescence showed that renal-CD100 protein was predominantly in tubules, while plexin-B1 was present in both glomeruli and tubules. To determine whether glomerular plexin-B1 mediates leucocyte recruitment via leucocyte CD100, recruitment was studied after passive transfer of heterologous antibody (attracting neutrophils) or isologous antibody (attracting macrophages). Glomerular macrophages were reduced in CD100(-/-) mice, but neutrophil recruitment was equivalent, consistent with CD100 expression on macrophages, but not neutrophils. CD100 promotes severe nephritogenic immune responses and leucocyte CD100-glomerular plexin-B1 interactions enhance macrophage recruitment to glomeruli.


Subject(s)
Antigens, CD/immunology , Glomerulonephritis/immunology , Kidney Glomerulus/immunology , Macrophages/immunology , Semaphorins/immunology , Animals , Disease Progression , Glomerular Basement Membrane/immunology , Immunity, Cellular , Immunoglobulin G/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Polymerase Chain Reaction/methods , Receptors, Cell Surface/metabolism
17.
Nephrol Dial Transplant ; 24(10): 3024-32, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19465557

ABSTRACT

BACKGROUND: IL-1beta has the potential to promote progressive renal disease by effects on macrophage recruitment and activation or by effects mediated through tubular cell transforming growth factor (TGF)-beta production, previously demonstrated in vitro. METHODS: The in vivo roles of endogenous IL-1beta and its type I receptor (IL-1RI) in renal fibrosis were studied using wild-type C57BL/6 mice, IL-1beta(-/-) and IL-1RI(-/-) mice with unilateral ureteric obstruction. RESULTS: After 7 days, IL-1RI(-/-) mice (IL-1alpha and IL-1beta deficient) were protected from injury and collagen accumulation. IL-1beta(-/-) mice demonstrated some histological protection, but no reduction in alpha1(1) procollagen mRNA or biochemically measured collagen accumulation. Compared with obstructed kidneys from wild-type mice, TGF-beta1 mRNA was reduced in IL-1RI(-/-) mice (with trends to reduced TGF-beta2 and TGF-beta3). Expression of a downstream TGF-beta effector, connective tissue growth factor, was decreased in IL-1RI(-/-) mice. IL-1RI(-/-) mice exhibited less tubulointerstitial apoptosis compared with wild-type mice. Macrophage infiltration and adhesion molecule mRNA expression was unchanged in IL-1beta(-/-) or IL-1RI(-/-) mice. While TNF expression was similar to wild-type mice, IFN-gamma expression was reduced in both IL-1beta(-/-) and IL-1RI(-/-) mice. IL-1RI(-/-) mice at 14 days showed a catch-up in fibrosis compared with wild-type mice. CONCLUSION: IL-1/IL-1RI interactions are profibrotic in renal fibrosis. IL-1RI(-/-) mice were more protected at an early stage, associated with changes in TGF-beta and downstream mediators of fibrosis, but independent of the presence of infiltrating macrophages.


Subject(s)
Kidney/pathology , Receptors, Interleukin-1 Type I/deficiency , Animals , Fibrosis/etiology , Mice , Mice, Inbred C57BL , Receptors, Interleukin-1 Type I/physiology
18.
J Am Soc Nephrol ; 19(3): 477-85, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18235099

ABSTRACT

T-bet is a transcription factor that is essential for T helper (Th)1 lineage commitment and optimal IFN-gamma production by CD4(+) T cells. We examined the role of T-bet in the development of experimental crescentic glomerulonephritis, which is induced by Th1-predominant, delayed-type hypersensitivity-like responses directed against a nephritogenic antigen. Anti-glomerular basement membrane (GBM) glomerulonephritis was induced in T-bet(-/-) and wild-type C57BL/6 mice. Compared with wild-type controls, renal injury was attenuated in T-bet(-/-) mice with glomerulonephritis, evidenced by less proteinuria, glomerular crescents, and tubulointerstitial inflammation. Accumulation of glomerular CD4(+) T cells and macrophages was decreased, and was associated with reduced intrarenal expression of the potent Th1 chemoattractants CCL5/RANTES and CXCL9/Mig. Supporting the pro-inflammatory nature of T-bet signaling, assessment of systemic immunity confirmed that T-bet(-/-) mice had a reduction in Th1 immunity. The kinetic profile of T-bet mRNA in wild-type mice supported the hypothesis that T-bet deficiency attenuates renal injury in part by shifting the Th1/Th2 balance away from a Th1 phenotype. Expression of renal and splenic IL-17A, characteristically expressed by the Th17 subset of effector T cells, which have been implicated in the pathogenesis of autoimmune disease, was increased in T-bet(-/-) mice. We conclude that T-bet directs Th1 responses that induce renal injury in experimental crescentic glomerulonephritis.


Subject(s)
Anti-Glomerular Basement Membrane Disease/metabolism , Interleukin-17/metabolism , Kidney/pathology , T-Box Domain Proteins/metabolism , Th1 Cells/physiology , Animals , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/pathology , Antigens, CD19/metabolism , Apoptosis/physiology , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Cell Proliferation , Cytokines/metabolism , Kidney/metabolism , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger/metabolism , Spleen/metabolism , Th1 Cells/metabolism
19.
J Immunol ; 177(5): 3406-12, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16920982

ABSTRACT

CD100, a member of the semaphorin family, is a costimulatory molecule in adaptive immune responses by switching off CD72's negative signals. However, CD100's potential pathogenetic effects in damaging immune responses remain largely unexplored. We tested the hypothesis that CD100 plays a pathogenetic role in experimental immune complex glomerulonephritis. Daily injection of horse apoferritin for 14 days induced immune complex formation, mesangial proliferative glomerulonephritis and proteinuria in CD100-intact (CD100+/+) BALB/c mice. CD100-deficient (CD100-/-) mice were protected from histological and functional glomerular injury. They exhibited reduced deposition of Igs and C3 in glomeruli, reduced MCP-1 and MIP-2 intrarenal mRNA expression, and diminished glomerular macrophage accumulation. Attenuated glomerular injury was associated with decreased Ag-specific Ig production, reduced CD4+ cell activation and cytokine production. Following Ag injection, CD4+ cell CD100 expression was enhanced and dendritic cell CD86 expression was up-regulated. However, in CD100-/- mice, dendritic cell CD86 (but not CD80) up-regulation was significantly attenuated. Following i.p. immunization, CD86, but not CD80, promotes early Ag-specific TCR-transgenic DO11.10 CD4+ cell proliferation and IFN-gamma production, suggesting that CD100 expression enables full expression of CD86 and consequent CD4+ cell activation. Transfer of CD100+/+ DO11.10 cells into CD100-/- mice resulted in decreased proliferation demonstrating that CD100 from other sources in addition to CD100 from Ag-specific CD4+ cells plays a role in initial T cell proliferation. Although T cell-B cell interactions also may be relevant, these studies demonstrate that CD100 enhances pathogenetic humoral immune responses and promotes the activation of APCs by up-regulating CD86 expression.


Subject(s)
Antigens, CD/immunology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Lymphocyte Activation/immunology , Semaphorins/immunology , Semaphorins/metabolism , Animals , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Survival , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/metabolism , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Immunization , Immunoglobulins/immunology , Injections, Intraperitoneal , Kidney/immunology , Kidney/injuries , Male , Mice , Mice, Knockout , Semaphorins/deficiency , Semaphorins/genetics
20.
Diabetes ; 54(2): 434-42, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15677501

ABSTRACT

The nature of the T-cell response to antigen is governed by the activation state of the antigen-presenting dendritic cell (DC). Immature or resting DCs have been shown to induce T-cell responses that may protect against the development of autoimmune disease. Effectively harnessing this "tolerogenic" effect of resting DCs requires that it be disease-specific and that activation of DCs by manipulation ex vivo is avoided. We reasoned that this could be achieved by transferring in vivo partially differentiated myeloid progenitor cells encoding a disease-specific autoantigen. With the aim of preventing autoimmune diabetes, we transferred myeloid progenitor cells encoding proinsulin into NOD mice. Bone marrow (BM) was cultured in granulocyte macrophage colony-stimulating factor (GM-CSF) and transforming growth factor-beta1, a cytokine combination that expands myeloid cells but inhibits terminal DC differentiation, to yield Gr-1(+)/CD11b(+)/CD11c(-) myeloid progenitor cells and a minor population of CD11c(+)/CD11b(+)/CD86(lo) immature DCs. After transfer, Gr-1(+) myeloid cells acquired the characteristics of resting DCs (CD11c(+)/MHC classII(int)/CD86(lo)/CD40(lo)). Gr-1(+) myeloid cells generated from transgenic NOD mice that expressed proinsulin controlled by a major histocompatibility complex (MHC) class II promoter, but not from wild-type NOD mice, transferred into 4-week-old female NOD mice significantly suppressed diabetes development. The transfer of DC progenitors encoding a disease-specific autoantigen is, therefore, an effective immunotherapeutic strategy that could be applied to humans.


Subject(s)
Dendritic Cells/cytology , Diabetes Mellitus, Type 1/prevention & control , Myeloid Cells/cytology , Proinsulin/genetics , Stem Cells/cytology , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Differentiation , Cells, Cultured , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immune Tolerance , Mice , Mice, Inbred NOD , Myeloid Cells/drug effects , Myeloid Cells/physiology , Stem Cells/drug effects , Stem Cells/physiology , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...