Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Am Soc Nephrol ; 27(6): 1635-49, 2016 06.
Article in English | MEDLINE | ID: mdl-26567242

ABSTRACT

Endothelial dysfunction is a central pathomechanism in diabetes-associated complications. We hypothesized a pathogenic role in this dysfunction of cathepsin S (Cat-S), a cysteine protease that degrades elastic fibers and activates the protease-activated receptor-2 (PAR2) on endothelial cells. We found that injection of mice with recombinant Cat-S induced albuminuria and glomerular endothelial cell injury in a PAR2-dependent manner. In vivo microscopy confirmed a role for intrinsic Cat-S/PAR2 in ischemia-induced microvascular permeability. In vitro transcriptome analysis and experiments using siRNA or specific Cat-S and PAR2 antagonists revealed that Cat-S specifically impaired the integrity and barrier function of glomerular endothelial cells selectively through PAR2. In human and mouse type 2 diabetic nephropathy, only CD68(+) intrarenal monocytes expressed Cat-S mRNA, whereas Cat-S protein was present along endothelial cells and inside proximal tubular epithelial cells also. In contrast, the cysteine protease inhibitor cystatin C was expressed only in tubules. Delayed treatment of type 2 diabetic db/db mice with Cat-S or PAR2 inhibitors attenuated albuminuria and glomerulosclerosis (indicators of diabetic nephropathy) and attenuated albumin leakage into the retina and other structural markers of diabetic retinopathy. These data identify Cat-S as a monocyte/macrophage-derived circulating PAR2 agonist and mediator of endothelial dysfunction-related microvascular diabetes complications. Thus, Cat-S or PAR2 inhibition might be a novel strategy to prevent microvascular disease in diabetes and other diseases.


Subject(s)
Cathepsins/physiology , Diabetic Angiopathies/etiology , Endothelial Cells/enzymology , Receptor, PAR-2/metabolism , Animals , Cathepsins/antagonists & inhibitors , Cells, Cultured , Kidney Glomerulus/cytology , Male , Mice , Microvessels , Proline/analogs & derivatives , Proline/pharmacology , Urothelium/cytology
2.
Am J Pathol ; 185(4): 1156-66, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25680278

ABSTRACT

Chronic renal disease (CRD) accelerates the development of atherosclerosis. The potent protease cathepsin S cleaves elastin and generates bioactive elastin peptides, thus promoting vascular inflammation and calcification. We hypothesized that selective cathepsin S inhibition attenuates atherogenesis in hypercholesterolemic mice with CRD. CRD was induced by 5/6 nephrectomy in high-fat high-cholesterol fed apolipoprotein E-deficient mice. CRD mice received a diet admixed with 6.6 or 60 mg/kg of the potent and selective cathepsin S inhibitor RO5444101 or a control diet. CRD mice had significantly higher plasma levels of osteopontin, osteocalcin, and osteoprotegerin (204%, 148%, and 55%, respectively; P < 0.05), which were inhibited by RO5444101 (60%, 40%, and 36%, respectively; P < 0.05). Near-infrared fluorescence molecular imaging revealed a significant reduction in cathepsin activity in treated mice. RO5444101 decreased osteogenic activity. Histologic assessment in atherosclerotic plaque demonstrated that RO5444101 reduced immunoreactive cathepsin S (P < 0.05), elastin degradation (P = 0.01), plaque size (P = 0.01), macrophage accumulation (P < 0.01), growth differentiation factor-15 (P = 0.0001), and calcification (alkaline phosphatase activity, P < 0.01; osteocalcin, P < 0.05). Furthermore, cathepsin S inhibitor or siRNA significantly decreased expression of growth differentiation factor-15 and monocyte chemotactic protein-1 in a murine macrophage cell line and human primary macrophages. Systemic inhibition of cathepsin S attenuates the progression of atherosclerotic lesions in 5/6 nephrectomized mice, serving as a potential treatment for atherosclerosis in patients with CRD.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/pathology , Cathepsins/antagonists & inhibitors , Kidney Failure, Chronic/enzymology , Kidney Failure, Chronic/pathology , Animals , Arteries/enzymology , Arteries/pathology , Atherosclerosis/complications , Biomarkers/blood , Cathepsins/metabolism , Chemokine CCL2/metabolism , Growth Differentiation Factor 15/metabolism , Humans , Interferon-gamma/pharmacology , Kidney Failure, Chronic/blood , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Osteogenesis/drug effects , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Vascular Calcification/complications , Vascular Calcification/pathology
3.
Retina ; 36(12): 2311-2318, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27648638

ABSTRACT

PURPOSE: To analyze the levels of lysophosphatidic acids (LPAs) and autotaxin (ATX) in undiluted vitreous of untreated patients with retinal vein occlusion (RVO). METHODS: Sixty-four vitreous samples (40 RVO, 24 controls with idiopathic floaters) were analyzed in this retrospective case series using LC/MS for LPAs 16:0, 18:0, 18:1, 20:4, and ELISA kits or Luminex technology for ATX, angiopoetin-1 (ANG-1), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin-8 (IL-8), monocyte chemoattractant protein-1 (MCP-1), pigment epithelium-derived factor (PEDF), and vascular endothelial growth factor (VEGF). LPA and ATX levels were correlated with the visual acuity, central macular thickness (CMT), average retinal thickness (AvT), vitreal cytokine levels and with each other. RESULTS: Levels of every LPA species tested and ATX were significantly increased in the vitreous fluid from all patients with RVO (total LPAs: 968.0 ± 842.3 nM; ATX: 2.5 ± 1.02 nM) compared with controls (total LPAs: 225.2 ± 292.8 nM, P < 0.0001; ATX: 1.9 ± 1.00 nM, P = 0.005). There were strong positive correlations between the vitreal levels of IL-6, IL-8, MCP-1, VEGF and LPAs. CONCLUSION: Levels of LPAs and ATX were positively correlated with proinflammatory cytokines and VEGF and might thus play an important role in the development of macular edema secondary to RVO.


Subject(s)
Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Retinal Vein Occlusion/metabolism , Aged , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Retrospective Studies , Vitreous Body/metabolism
4.
Ann Rheum Dis ; 74(2): 452-63, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24300027

ABSTRACT

OBJECTIVES: Major histocompatibility complex (MHC) class II-mediated priming of T and B lymphocytes is a central element of autoimmunity in systemic lupus erythematosus (SLE) and lupus nephritis. The cysteine protease cathepsin S degrades the invariant peptide chain during MHC II assembly with antigenic peptide in antigen-presenting cells; therefore, we hypothesised that cathepsin S inhibition would be therapeutic in SLE. METHODS: We developed a highly specific small molecule, orally available, cathepsin S antagonist, RO5461111, with suitable pharmacodynamic and pharmacokinetic properties that efficiently suppressed antigen-specific T cell and B cell priming in vitro and in vivo. RESULTS: When given to MRL-Fas(lpr) mice with SLE and lupus nephritis, RO5461111 significantly reduced the activation of spleen dendritic cells and the subsequent expansion and activation of CD4 T cells and CD4/CD8 double-negative T cells. Cathepsin S inhibition impaired the spatial organisation of germinal centres, suppressed follicular B cell maturation to plasma cells and Ig class switch. This reversed hypergammaglobulinemia and significantly suppressed the plasma levels of numerous IgG (but not IgM) autoantibodies below baseline, including anti-dsDNA. This effect was associated with less glomerular IgG deposits, which protected kidneys from lupus nephritis. CONCLUSIONS: Together, cathepsin S promotes SLE by driving MHC class II-mediated T and B cell priming, germinal centre formation and B cell maturation towards plasma cells. These afferent immune pathways can be specifically reversed with the cathepsin S antagonist RO5461111, which prevents lupus nephritis progression even when given after disease onset. This novel therapeutic strategy could correct a common pathomechanism of SLE and other immune complex-related autoimmune diseases.


Subject(s)
Cathepsins/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Lymphocyte Activation/drug effects , Proline/analogs & derivatives , Animals , B-Lymphocytes/immunology , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Proline/pharmacology , Real-Time Polymerase Chain Reaction
5.
Diabetologia ; 57(8): 1674-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24891017

ABSTRACT

AIMS/HYPOTHESIS: Cathepsin S (CatS) belongs to a family of proteases that have been implicated in several disease processes. We previously identified CatS as a protein that is markedly overexpressed in adipose tissue of obese individuals and downregulated after weight loss and amelioration of glycaemic status induced by gastric bypass surgery. This prompted us to test whether the protease contributes to the pathogenesis of type 2 diabetes using mouse models with CatS inactivation. METHODS: CatS knockout mice and wild-type mice treated with orally active small-molecule CatS inhibitors were fed chow or high-fat diets and explored for change in glycaemic status. RESULTS: CatS deletion induced a robust reduction in blood glucose, which was preserved in diet-induced obesity and with ageing and was recapitulated with CatS inhibition in obese mice. In vivo testing of glucose tolerance, insulin sensitivity and glycaemic response to gluconeogenic substrates revealed that CatS suppression reduced hepatic glucose production despite there being no improvement in insulin sensitivity. This phenotype relied on downregulation of gluconeogenic gene expression in liver and a lower rate of hepatocellular respiration. Mechanistically, we found that the protein 'regulated in development and DNA damage response 1' (REDD1), a factor potentially implicated in reduction of respiratory chain activity, was overexpressed in the liver of mice with CatS deficiency. CONCLUSIONS/INTERPRETATION: Our results revealed an unexpected metabolic effect of CatS in promoting pro-diabetic alterations in the liver. CatS inhibitors currently proposed for treatment of autoimmune diseases could help to lower hepatic glucose output in obese individuals at risk for type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Cathepsins/antagonists & inhibitors , Cathepsins/genetics , Insulin Resistance/physiology , Obesity/metabolism , Animals , Cathepsins/metabolism , Diet, High-Fat , Insulin/metabolism , Mice , Mice, Knockout , Oxygen Consumption/physiology
6.
Am J Emerg Med ; 32(1): 82-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24079985

ABSTRACT

The current resuscitation guidelines of the European Resuscitation Council do not include automatic chest compression devices (ACDs) as standard equipment to support cardiopulmonary resuscitation attempts. One possible reason could be the lack of a list of indications and contraindications for the use of ACD systems. This review should give a summary of current studies and developments according to ACD systems and deliver a list of possible applications. Furthermore, we discuss some ethical problems with cardiopulmonary resuscitation attempts and, in particular, with ACD systems. The use of ACDs occurs instead of manual chest compression. Because of this, there is no reason for changing the current guidelines, especially termination recommendations while using ACD systems. From our point of view, ACDs are a very good supplement to the current standard of resuscitation according to the European Resuscitation Council guidelines.


Subject(s)
Cardiopulmonary Resuscitation/instrumentation , Heart Massage/instrumentation , Contraindications , Heart Arrest/therapy , Humans , Practice Guidelines as Topic
7.
bioRxiv ; 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38895439

ABSTRACT

Lysosomes catabolize lipids and other biological molecules, a function essential for cellular and organismal homeostasis. Key to lipid catabolism in the lysosome is bis(monoacylglycero)phosphate (BMP), a major lipid constituent of intralysosomal vesicles (ILVs) and a stimulator of lipid-degrading enzymes. BMP levels are altered in a broad spectrum of human conditions, including neurodegenerative diseases. Although BMP synthase was recently discovered, it has long been thought that BMP's unique stereochemistry confers resistance to acid phospholipases, a requirement for its role in the lysosome. Here, we demonstrate that PLA2G15, a major lysosomal phospholipase, efficiently hydrolyzes BMP with primary esters regardless of stereochemistry. Interestingly, we discover that BMP's unique esterification position is what confers resistance to hydrolysis. Purified PLA2G15 catabolizes most BMP species derived from cell and tissue lysosomes under acidic conditions. Furthermore, PLA2G15 catalytic activity against synthesized BMP stereoisomers with primary esters was comparable to its canonical substrates. Conversely, BMP with secondary esters is intrinsically stable in vitro and requires acyl migration for hydrolysis in lysosomes. Consistent with our biochemical data, PLA2G15-deficient tissues and cells accumulate multiple BMP species, a phenotype reversible by supplementing wildtype PLA2G15 but not its catalytically dead mutant. Increasing BMP levels by targeting PLA2G15 reverses the cholesterol accumulation phenotype in Niemann Pick Disease Type C (NPC1) patient fibroblasts and significantly ameliorate disease pathologies in NPC1-deficient mice leading to extended lifespan. Our findings establish the rules that govern the stability of BMP in the lysosome and identify PLA2G15 as a lysosomal BMP hydrolase and as a potential target for modulating BMP levels for therapeutic intervention.

8.
J Pathol ; 228(2): 131-47, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22570261

ABSTRACT

The incidence of chronic kidney diseases (CKD) is constantly rising, reaching epidemic proportions in the western world and leading to an enormous threat, even to modern health-care systems, in industrialized countries. Therapies of CKD have greatly improved following the introduction of drugs targeting the renin-angiotensin system (RAAS) but even this refined pharmacological approach has failed to stop progression to end-stage renal disease (ESRD) in many individuals. In vitro historical data and recent new findings have suggested that progression of renal fibrosis might occur as a result of an altered tubulo-interstitial microenvironment and, more specifically, as a result of an altered epithelial-mesenchymal crosstalk. Here we the review biological findings that support the hypothesis of an altered cellular crosstalk in an injured local tubulo-interstitial microenvironment leading to renal disease progression. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Communication/physiology , Epithelial-Mesenchymal Transition/physiology , Kidney/pathology , Cellular Microenvironment , Disease Progression , Epithelial Cells/pathology , Feedback, Physiological/physiology , Fibrosis , Humans , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/physiopathology , Kidney Tubules/pathology , Mesoderm/pathology , Renin-Angiotensin System/physiology
9.
Sci Rep ; 13(1): 18550, 2023 10 29.
Article in English | MEDLINE | ID: mdl-37899458

ABSTRACT

Neuronal ceroid lipofuscinosis 6 (CLN6) is a rare and fatal autosomal recessive disease primarily affecting the nervous system in children. It is caused by a pathogenic mutation in the CLN6 gene for which no therapy is available. Employing an untargeted metabolomics approach, we analyzed the metabolic changes in CLN6 subjects to see if this system could potentially yield biomarkers for diagnosis and monitoring disease progression. Neuronal-like cells were derived from human fibroblast lines from CLN6-affected subjects (n = 3) and controls (wild type, n = 3). These were used to assess the potential of a neuronal-like cell-based metabolomics approach to identify CLN6 distinctive and specific biomarkers. The most impacted metabolic profile is associated with sphingolipids, glycerophospholipids metabolism, and calcium signaling. Over 2700 spectral features were screened, and fifteen metabolites were identified that differed significantly between both groups, including the sphingolipids C16 GlcCer, C24 GlcCer, C24:1 GlcCer and glycerophospholipids PG 40:6 and PG 40:7. Of note, these fifteen metabolites were downregulated in the CLN6 disease group. This study is the first to analyze the metabolome of neuronal-like cells with a pathogenic mutation in the CLN6 gene and to provide insights into their metabolomic alterations. This could allow for the development of novel biomarkers for monitoring CLN6 disease.


Subject(s)
Membrane Proteins , Neuronal Ceroid-Lipofuscinoses , Child , Humans , Membrane Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Lipid Metabolism , Metabolomics , Glycerophospholipids , Sphingolipids , Biomarkers/metabolism
10.
Nephrol Dial Transplant ; 27 Suppl 3: iii43-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22785113

ABSTRACT

Observational clinical studies link acute kidney injury to chronic kidney disease (CKD) progression. The pathophysiological mechanisms that underlie this process are currently unknown but recently published papers suggest that tubular epithelial cells and interstitial mesenchymal cells emerge as a single unit, and their integrity alteration as a whole might lead to renal fibrosis and CKD. The present article reviews the biological findings supporting the hypothesis of an altered epithelial/mesenchymal crosstalk in fibrosis development and progression toward CKD.


Subject(s)
Acute Kidney Injury/physiopathology , Epithelial-Mesenchymal Transition , Kidney Failure, Chronic/physiopathology , Disease Progression , Humans
11.
Nephrol Dial Transplant ; 27 Suppl 3: iii81-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22734108

ABSTRACT

Chronic kidney disease (CKD) and end-stage renal disease (ESRD) are currently considered as major health burdens. Notably, CKD can be regarded as an interesting clinical model of accelerated cardiovascular disease (CVD) and ageing, which offers exciting new perspectives and challenges for pharmaceutical drug development. However, during the last decades, therapeutic advances to slow down the progression of CKD and reduce CVD risk have largely failed due to several possible reasons including (i) the lack of profound understanding of the pathophysiology of chronic renal damage and its associated CVD; (ii) an inadequate characterization of molecular mechanisms of currently approved therapies such as renin-angiotensin-aldosterone-system (RAAS) blockade; (iii) the unclear biochemical property needs required for novel therapeutic approaches; (iv) the missing quantity and quality of clinical trials in the nephrology field; and, most importantly, (v) the absence of prognostic renal biomarkers that reflect the severity of the structural organ damage and predict ESRD as well as CVD mortality. There is clearly an insufficient understanding of why a significant proportion of CKD patients progress to ESRD and/or die from CVD whereas others rather remain stable. In this article, we urge renal researchers to develop novel experimental and clinical tools for rational and translational drug discovery. Identification of individualized determinants of CKD progression and/or premature CVD will enable personalized medicine and lead to novel innovative nephro- and/or cardioprotective pharmacological treatment in these high-risk patients.


Subject(s)
Biomarkers/analysis , Cardiovascular Diseases/diagnosis , Drug Design , Drug Discovery/trends , Precision Medicine , Renal Insufficiency, Chronic/diagnosis , Research Design/trends , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Disease Progression , Humans , Prognosis , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy
12.
J Immunol ; 184(8): 4568-74, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20237293

ABSTRACT

T lymphocytes of the Th2 type are central orchestrators of airway inflammation in asthma. The mechanisms that regulate their accumulation in the asthmatic airways remains poorly understood. We tested the hypothesis that CCR4, preferentially expressed on T lymphocytes of the Th2 type, plays a critical role in this process. We enumerated by flow cytometry the CCR4-expressing T cells from blood, induced sputum, and biopsy samples of patients with asthma and control subjects. We showed a positive correlation between the numbers of peripheral blood CCR4+ T cells and asthma severity, provided evidence of preferential accumulation of CCR4+ T cells in asthmatic airways, and demonstrated that CCR4+ but not CCR4- cells from patients with asthma produce Th2 cytokines. Explanted airway mucosal biopsy specimens, acquired by bronchoscopy from subjects with asthma, were challenged with allergen and the explant supernatants assayed for T cell chemotactic activity. Allergen-induced ex vivo production of the CCR4 ligand, CCL17 was raised in explants from patients with asthma when compared with healthy controls. Using chemotaxis assays, we showed that the T cell chemotactic activity generated by bronchial explants can be blocked with a selective CCR4 antagonist or by depleting CCR4+ cells from responder cells. These results provide evidence that CCR4 might play a role in allergen-driven Th2 cell accumulation in asthmatic airways. Targeting this chemokine receptor in patients with asthma might reduce Th2 cell-driven airway inflammation; therefore, CCR4 antagonists could be an effective new therapy for asthma. This study also provides wider proof of concept for using tissue explants to study immunomodulatory drugs for asthma.


Subject(s)
Asthma/immunology , Asthma/pathology , Chemotaxis, Leukocyte/immunology , Lung/immunology , Lung/pathology , Receptors, CCR4/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Animals , Antigens, Dermatophagoides/immunology , Arthropod Proteins , Asthma/metabolism , Bronchi/cytology , Bronchi/immunology , Bronchi/metabolism , Chronic Disease , Cysteine Endopeptidases , Cytokines/biosynthesis , Dermatophagoides pteronyssinus/immunology , Humans , Lung/metabolism , Pilot Projects , Receptors, CCR4/antagonists & inhibitors , Receptors, CCR4/biosynthesis , Receptors, CCR4/blood , Severity of Illness Index , T-Lymphocyte Subsets/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Up-Regulation/immunology
13.
Front Neurol ; 13: 907317, 2022.
Article in English | MEDLINE | ID: mdl-35734474

ABSTRACT

Gaucher Disease (GD) 2 is a rare inherited lysosomal disorder. Early-onset and rapid progression of neurovisceral symptoms lead to fatal outcome in early childhood. Treatment is symptomatic, a curative therapy is currently not available. This prospective study describes the clinical and biochemical outcome of a GD 2 patient treated with high dose ambroxol from the age of 4 months. Due to progressive hepatosplenomegaly additional enzyme replacement therapy was required 1 year after ambroxol monotherapy was initiated. Detailed clinical follow-up data demonstrated an age-appropriate neurocognitive and motor development but no clear benefit on peripheral organs. Glucosylsphingosine (Lyso-GL1) in cerebrospinal fluid decreased remarkably compared to pre-treatment, whereas Lyso-GL1 and chitotriosidase in blood increased. Ambroxol treatment of patient fibroblasts revealed a significant increase in ß-glucocerebrosidase activity in vitro. To our knowledge, this is the first report of a GD 2 patient with age-appropriate cognitive and motor development at 3 years of age. Combination of high dose ambroxol with ERT proved to be a successful approach to manage both visceral and neurological manifestations.

14.
Kidney Int ; 80(1): 68-78, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21508925

ABSTRACT

The progression of diabetic nephropathy is associated with an infiltration of macrophages expressing different phenotypes. As classically activated chemokine receptor CCR2+ macrophages are thought to drive tissue inflammation and remodeling, we tested whether blocking CCR2 could reduce intrarenal inflammation and prevent glomerulosclerosis in type 2 diabetes. This was achieved with RO5234444, an orally active small-molecule CCR2 antagonist that blocks ligand binding, its internalization, and monocyte chemotaxis. Male type 2 diabetic db/db mice were uninephrectomized to increase glomerular hyperfiltration to accelerate the development of glomerulosclerosis. From 16 weeks until killing at 24 weeks of age, mice were chow fed with or without admixed antagonist to achieve a trough plasma concentration above IC50 for binding in the mouse. CCR2 blockade reduced circulating monocyte levels, but did not affect total leukocyte or neutrophil numbers, and was associated with a reduction in the number of macrophages and apoptotic podocytes in the glomerulus. This treatment resulted in a higher total number of podocytes, less glomerulosclerosis, reduced albuminuria, and a significantly improved glomerular filtration rate. This successful pre-clinical trial suggests that this antagonist may now be ready for testing in humans with the nephropathy of diabetes mellitus.


Subject(s)
Cinnamates/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/prevention & control , Piperazines/pharmacology , Receptors, CCR2/antagonists & inhibitors , Renal Insufficiency/prevention & control , Administration, Oral , Albuminuria/drug therapy , Animals , Cinnamates/administration & dosage , Cinnamates/chemistry , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Diabetic Nephropathies/pathology , Glomerular Filtration Rate/drug effects , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Piperazines/administration & dosage , Piperazines/chemistry , Podocytes/drug effects , Podocytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR2/genetics
15.
PLoS One ; 15(12): e0244368, 2020.
Article in English | MEDLINE | ID: mdl-33370388

ABSTRACT

Achondroplasia is a rare genetic disorder caused by mutations in the Fibroblast Growth Factor receptor 3 (FGFR3). These mutations lead to aberrant increase of inhibitory signaling in proliferating chondrocytes at the growth plate. Recifercept is a potential treatment for this disease using a decoy approach to sequester FGFR3 ligands subsequently normalizing activation of the mutated FGFR3 receptor. Recifercept binds to FGF isoforms in vitro and in cellular model systems and reduces FGFR3 signaling. In addition, in a transgenic mouse model of achondroplasia, Recifercept restores reduced body weight and long bone growth in these mice. These data suggest that Recifercept treatment could lead to clinical benefits in children treated with this molecule.


Subject(s)
Achondroplasia/drug therapy , Fibroblast Growth Factors/metabolism , Mutation , Receptor, Fibroblast Growth Factor, Type 3/administration & dosage , Achondroplasia/genetics , Achondroplasia/metabolism , Animals , Body Weight/drug effects , Bone Development/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Protein Binding/drug effects , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 3/pharmacology , Signal Transduction/drug effects
16.
Sci Rep ; 7(1): 2775, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28584258

ABSTRACT

Cathepsin(Cat)-S processing of the invariant chain-MHC-II complex inside antigen presenting cells is a central pathomechanism of autoimmune-diseases. Additionally, Cat-S is released by activated-myeloid cells and was recently described to activate protease-activated-receptor-(PAR)-2 in extracellular compartments. We hypothesized that Cat-S blockade targets both mechanisms and elicits synergistic therapeutic effects on autoimmune tissue injury. MRL-(Fas)lpr mice with spontaneous autoimmune tissue injury were treated with different doses of Cat-S inhibitor RO5459072, mycophenolate mofetil or vehicle. Further, female MRL-(Fas)lpr mice were injected with recombinant Cat-S with/without concomitant Cat-S or PAR-2 blockade. Cat-S blockade dose-dependently reversed aberrant systemic autoimmunity, e.g. plasma cytokines, activation of myeloid cells and hypergammaglobulinemia. Especially IgG autoantibody production was suppressed. Of note (MHC-II-independent) IgM were unaffected by Cat-S blockade while they were suppressed by MMF. Cat-S blockade dose-dependently suppressed immune-complex glomerulonephritis together with a profound and early effect on proteinuria, which was not shared by MMF. In fact, intravenous Cat-S injection induced severe glomerular endothelial injury and albuminuria, which was entirely prevented by Cat-S or PAR-2 blockade. In-vitro studies confirm that Cat-S induces endothelial activation and injury via PAR-2. Therapeutic Cat-S blockade suppresses systemic and peripheral pathomechanisms of autoimmune tissue injury, hence, Cat-S is a promising therapeutic target in lupus nephritis.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/pathology , Autoimmunity/drug effects , Cathepsins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Animals , Autoimmune Diseases/drug therapy , Cathepsins/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacokinetics , Female , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/drug therapy , Lupus Nephritis/etiology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Mice , Mice, Inbred MRL lpr , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism
18.
EMBO Mol Med ; 8(11): 1265-1288, 2016 11.
Article in English | MEDLINE | ID: mdl-27742718

ABSTRACT

Anti-angiogenic therapies using biological molecules that neutralize vascular endothelial growth factor-A (VEGF-A) have revolutionized treatment of retinal vascular diseases including age-related macular degeneration (AMD). This study reports preclinical assessment of a strategy to enhance anti-VEGF-A monotherapy efficacy by targeting both VEGF-A and angiopoietin-2 (ANG-2), a factor strongly upregulated in vitreous fluids of patients with retinal vascular disease and exerting some of its activities in concert with VEGF-A. Simultaneous VEGF-A and ANG-2 inhibition was found to reduce vessel lesion number, permeability, retinal edema, and neuron loss more effectively than either agent alone in a spontaneous choroidal neovascularization (CNV) model. We describe the generation of a bispecific domain-exchanged (crossed) monoclonal antibody (CrossMAb; RG7716) capable of binding, neutralizing, and depleting VEGF-A and ANG-2. RG7716 showed greater efficacy than anti-VEGF-A alone in a non-human primate laser-induced CNV model after intravitreal delivery. Modification of RG7716's FcRn and FcγR binding sites disabled the antibodies' Fc-mediated effector functions. This resulted in increased systemic, but not ocular, clearance. These properties make RG7716 a potential next-generation therapy for neovascular indications of the eye.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Antibodies, Monoclonal/administration & dosage , Eye Diseases/drug therapy , Immunologic Factors/administration & dosage , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Eye Diseases/pathology , Immunologic Factors/isolation & purification , Immunologic Factors/pharmacology , Macaca fascicularis , Treatment Outcome
20.
J Med Chem ; 56(23): 9789-801, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24224654

ABSTRACT

Starting from the weakly active dual CatS/K inhibitor 5, structure-based design supported by X-ray analysis led to the discovery of the potent and selective (>50,000-fold vs CatK) cyclopentane derivative 22 by exploiting specific ligand-receptor interactions in the S2 pocket of CatS. Changing the central cyclopentane scaffold to the analogous pyrrolidine derivative 57 decreased the enzyme as well as the cell-based activity significantly by 24- and 69-fold, respectively. The most promising scaffold identified was the readily accessible proline derivative (e.g., 79). This compound, with an appealing ligand efficiency (LE) of 0.47, included additional structural modifications binding in the S1 and S3 pockets of CatS, leading to favorable in vitro and in vivo properties. Compound 79 reduced IL-2 production in a transgenic DO10.11 mouse model of antigen presentation in a dose-dependent manner with an ED50 of 5 mg/kg.


Subject(s)
Cathepsins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemical synthesis , Animals , Cyclopentanes/chemistry , Cysteine Proteinase Inhibitors/pharmacokinetics , Humans , Mice , Proline/analogs & derivatives , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL