Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Biomedicines ; 8(11)2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33218034

ABSTRACT

Recent preclinical data show that sodium glucose cotransporter 2 (SGLT2) inhibitors are able to reduce weight gain and induce beiging in white adipose tissue (WAT). We have previously shown that in neurogenic hypertensive Schlager (BPH/2J) mice, treatment with the SGLT2 inhibitor, Dapagliflozin, reduced blood pressure and prevented weight gain. Here we show that chemical sympathetic denervation achieved by systemic administration of 6-hydroxy-dopamine (6-OHDA) reduces body weight and the heightened sympathetic nervous system (SNS) innervation in WAT. Furthermore, we demonstrate that 2 weeks of Dapagliflozin treatment increases SNS innervation in WAT of hypertensive mice. This increase is accompanied by a non-significant elevation in mRNA levels of the Ucp1 and Pgc-1α genes, which are markers of beiging. No significant difference in the mRNA levels of the inflammatory mediators Il-6 and Tnf-α were detected in WAT of Dapagliflozin treated mice. These findings suggest that SGLT-2 inhibitor-associated prevention of weight gain may be mediated, at least in part, by inducing the beiging of WAT.

2.
J Clin Med ; 9(9)2020 Aug 26.
Article in English | MEDLINE | ID: mdl-32858953

ABSTRACT

Studying the role of circulatory factors in the pathogenesis of diseases has been key to the development of effective therapies. We sought to examine the effect of antihypertensive therapies on numerous circulatory factors including short chain fatty acids and growth factors in a human cohort. A subset of participants from an earlier study was characterized by their hypertensive and/or treatment status and separated into three groups: (i) normotensives; (ii) untreated hypertensive and (iii) treated hypertensive subjects. Circulating levels of short chain fatty acids, FGF21 and TNF superfamily members were measured as part of this study. Both F2-isoprostane and circulating lipid levels were reanalysed as part of this current study. We found that antihypertensive treatment increased butyrate levels and decreased acetate levels to levels similar to normotensives. We also found that antihypertensive treatments reduced levels of circulating FGF21, TNFSF14 and TNF-α. In conclusion, we identified several circulatory factors that are altered in hypertension.

3.
Calcif Tissue Int ; 107(3): 230-239, 2020 09.
Article in English | MEDLINE | ID: mdl-32638038

ABSTRACT

Identification of variants in the calcium-sensing receptor (CASR) gene is an important means of distinguishing between familial hypocalciuric hypercalcaemia (FHH) and primary hyperparathyroidism. However, identification and bioinformatics analysis of genetic variants alone is now considered insufficient as definitive proof; additional functional assessment is required to diagnose FHH with certainty. We identified two novel variants, D433Y and C739Y, and one previously reported variant G509R in the CASR of four kindreds provisionally diagnosed with FHH and aimed to functionally characterise these variants to confirm the diagnosis. Variant receptors were cloned as FLAG-tagged constructs into the mammalian expression vector, pcDNA3.1. Wild type and variant receptor constructs were expressed in HEK293 cells and their expression assessed by Western blot analysis and their functionality analysed using an IP-One assay which measures myo-inositol 1-phosphate accumulation following CaSR activation. Western blot analysis showed that the D433Y receptor had diminished mature glycosylated receptor compared with wild type CaSR whereas the G509R receptor had a complete lack of mature receptor. The C739Y receptor was consistently overexpressed. Functional assessment showed the D433Y receptor to be mildly inactivating at physiological calcium concentrations whereas the G509R receptor was inactive at all calcium concentrations. By contrast, the C739Y variant was activating compared to wild type receptor which is inconsistent with it causing FHH. We conclude that functional assessment of CaSR variants using the IP-One assay was useful in the investigation of suspected FHH probands, confirming the D433Y and G509R variants as likely pathogenic/pathogenic, but dismissing the C739Y variant as causing FHH.


Subject(s)
Hypercalcemia , Receptors, Calcium-Sensing , Calcium , HEK293 Cells , Humans , Hypercalcemia/congenital , Hypercalcemia/genetics , Mutation , Receptors, Calcium-Sensing/genetics
4.
World J Gastroenterol ; 26(23): 3225-3235, 2020 Jun 21.
Article in English | MEDLINE | ID: mdl-32684737

ABSTRACT

BACKGROUND: Type 1 diabetes (T1D) is associated with major chronic microvascular complications which contribute significantly to diabetes associated morbidity. The protein primarily responsible for glucose reabsorption in the kidney is sodium glucose co-transporter 2 (SGLT2). Presently, SGLT2 inhibitors are widely used in diabetic patients to improve blood glucose levels and prevent cardiovascular and renal complications. Given the broad therapeutic application of SGLT2 inhibitors, we hypothesised that SGLT2 inhibition may exert its protective effects via alterations of the gut microbiome and tested this in a type 1 diabetic mouse model of diabetic retinopathy. AIM: To determine whether the treatment with two independent SGLT2 inhibitors affects gut health in a type 1 diabetic mouse model. METHODS: The SGLT2 inhibitors empagliflozin or dapagliflozin (25 mg/kg/d) or vehicle dimethylsulfoxide (DMSO) were administered to C57BL/6J, Akita, Kimba and Akimba mice at 10 wk of age for 8 wk via their drinking water. Serum samples were collected and the concentration of succinate and the short chain fatty acid (SCFA) butyric acid was measured using gas chromatography-mass spectrometry. Enzyme-linked immunosorbent assay (ELISA) was performed to determine the concentration of insulin and leptin. Furthermore, the norepinephrine content in kidney tissue was determined using ELISA. Pancreatic tissue was collected and stained with haematoxylin and eosin and analysed using brightfield microscopy. RESULTS: Due to the presence of the Akita allele, both Akita and Akimba mice showed a reduction in insulin production compared to C57BL/6J and Kimba mice. Furthermore, Akita mice also showed the presence of apoptotic bodies within the pancreatic islets. The acinar cells of Akita and Akimba mice showed swelling which is indicative of acute injury or pancreatitis. After 8 wk of SGLT2 inhibition with dapagliflozin, the intermediate metabolite of gut metabolism known as succinate was significantly reduced in Akimba mice when compared to DMSO treated mice. In addition, empagliflozin resulted in suppression of succinate levels in Akimba mice. The beneficial SCFA known as butyric acid was significantly increased in Akita mice after treatment with dapagliflozin when compared to vehicle treated mice. The norepinephrine content in the kidney was significantly reduced with both dapagliflozin and empagliflozin therapy in Akita mice and was significantly reduced in Akimba mice treated with empagliflozin. In non-diabetic C57BL/6J and Kimba mice, serum leptin levels were significantly reduced after dapagliflozin therapy. CONCLUSION: The inhibition of SGLT2 reduces the intermediate metabolite succinate, increases SCFA butyric acid levels and reduces norepinephrine content in mouse models of T1D. Collectively, these improvements may represent an important mechanism underlying the potential benefits of SGLT2 inhibition in T1D and its complications.


Subject(s)
Diabetes Mellitus, Experimental , Animals , Diabetes Mellitus, Experimental/drug therapy , Glucose , Humans , Mice , Mice, Inbred C57BL , Sodium , Sodium-Glucose Transporter 2 , Succinic Acid
5.
Expert Opin Pharmacother ; 21(10): 1157-1166, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32301361

ABSTRACT

INTRODUCTION: Sodium-glucose cotransporter 2 (SGLT2) inhibitors such as Empagliflozin are novel antihyperglycemic drugs approved for the treatment of type 2 diabetes (T2D). In addition to its glucose-lowering effects, Empagliflozin promotes weight loss, blood pressure reduction, and other beneficial metabolic benefits. AREAS COVERED: This review outlines the pharmacokinetics, pharmacodynamics, safety, and tolerability of Empagliflozin and discusses its role in diabetes-associated hypertension. EXPERT OPINION: Empagliflozin was the first in class to not only demonstrate safety of SGLT2 inhibition but also cardio- and reno-protective effects in an adequately powered cardiovascular outcome trial. The EMPA-REG study showed significant reductions in mortality from cardiovascular causes, hospitalization for heart failure, and progression of diabetic kidney disease. These benefits cannot be attributed to glycemic control alone, suggesting the involvement of other SGLT2 inhibition-mediated mechanisms. Recent data suggests the potential utility of SGLT2 inhibition in other conditions including type 1 diabetes (T1D) and non-diabetic heart failure patients with clinical trials currently being conducted. In concert with ongoing pre-clinical investigations to unravel the mechanisms contributing to cardiorenal protection, the full therapeutic potential of SGLT2 inhibition will become apparent over the next few years and promises to be one of the major success stories in clinical medicine. ABBREVIATIONS: T1D: type 1 diabetes; T2D: type 2 diabetes; SGLT2: sodium-glucose cotransporter 2; CVD: cardiovascular disease; SBP: systolic blood pressure; DBP: diastolic blood pressure; SNS: sympathetic nervous system; BP: blood pressure; CV: cardiovascular; ZDF: Zucker diabetic fatty; CKD: chronic kidney disease; FDA: Food and Drug Administration.


Subject(s)
Benzhydryl Compounds/therapeutic use , Glucosides/therapeutic use , Hypertension/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Humans , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
6.
JACC Basic Transl Sci ; 5(2): 169-179, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32140623

ABSTRACT

Recent clinical trial data suggest a cardiorenal protective effect of sodium glucose cotransporter 2 (SGLT2) inhibition. We demonstrate that chemical denervation in neurogenic hypertensive Schlager (BPH/2J) mice reduced blood pressure, improved glucose homeostasis, and reduced renal SGLT2 protein expression. Inhibition of SGLT2 prevented weight gain, reduced blood pressure, significantly reduced elevations of tyrosine hydroxylase and norepinephrine, and protects against endothelial dysfunction. These findings provide evidence for significant crosstalk between activation of the sympathetic nervous system and SGLT2 regulation and possible ancillary effects on endothelial function, which may contribute to the observed cardiorenal protective effects of SGLT2 inhibition.

7.
Neural Regen Res ; 15(3): 512-518, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31571663

ABSTRACT

Hypertension is a risk factor for a large number of vision-threatening eye disorders. In this study, we investigated for the first time the retinal neural structure of the hypertensive BPH/2J mouse (Schlager mouse) and compared it to its control counterpart, the normotensive BPN/3J strain. The BPH/2J mouse is a selectively inbred mouse strain that develops chronic hypertension due to elevated sympathetic nervous system activity. When compared to the BPN/3J strain, the hypertensive BPH/2J mice showed a complete loss of outer layers of the neural retina at 21 weeks of age, which was indicative of a severe vision-threatening disease potentially caused by hypertension. To elucidate whether the retinal neural phenotype in the BPH/2J strain was attributed to increased BP, we investigated the neural retina of both BPN/3J and BPH/2J mice at 4 weeks of age. Our preliminary results showed for the first time that the BPH/2J strain develops severe retinal neural damage at a young age. Our findings suggest that the retinal phenotype in the BPH/2J mouse is possibly due to elevated blood pressure and may be contributed by an early onset spontaneous mutation which is yet to be identified or a congenital defect occurring in this strain. Further characterization of the BPH/2J mouse strain is likely to i) elucidate gene defects underlying retinal disease; ii) understand mechanisms leading to neural retinal disease and iii) permit testing of molecules for translational research to interfere with the progression of retinal disease. The animal experiments were performed with the approval of the Royal Perth Hospital Animal Ethics Committee (R535/17-18) on June 1, 2017.

8.
Am J Ophthalmol ; 204: 113-123, 2019 08.
Article in English | MEDLINE | ID: mdl-30878487

ABSTRACT

PURPOSE: To assess the safety and the 3-year results of combined phase 1 and 2a randomized controlled trials of rAAV.sFLT-1 gene therapy (GT) for wet age-related macular degeneration. DESIGN: Phase 1/2a clinical trial. METHODS: Patients were prospectively randomized into control (n = 13) and GT (n = 24) groups. GT patients received 1X1011vg rAAV.sFLT-1 and were seen every month for 1 year then as needed every 1 to 2 months. They were given retreatment anti-vascular endothelial growth factor injections according to predetermined criteria. At 12 months, GT patients were divided into 2 groups: HD-1 (n = 14), requiring <2, and HD-2 (n = 10), requiring >2 retreatments. RESULTS: Between 1 year and 3 years there were 3 adverse events (AEs) and 33 serious AEs reported. Of these, 15 occurred in the 13 control subjects and 21 in the 24 GT patients. Except for 1 case of transient choroiditis in a control patient, serious AEs were deemed to be unrelated to the study. Control patients received a median of 7.0 retreatments and lost a median of 7.0 Early Treatment Diabetic Retinopathy Study (ETDRS) letters, HD-1 patients received a median of 2.5 retreatments and lost a median of 4.0 ETDRS letters, and HD-2 patients received a median of 11.0 retreatments and lost a median of 7.0 ETDRS letters over 3 years. Center point thickness fluctuated. Thirty-three percent of control subjects, 44% of HD-2 patients, and 51% of HD-1 patients showed maintenance of baseline visual acuity. Four HD-1 patients (34%) maintained significant visual improvement at 3 years. None of these observations were statistically significant. CONCLUSIONS: Given the small number of patients, this study was unable to unequivocally confirm the existence of a biologic efficacy signal; however, it confirmed that rAAV.sFLT-1 gene delivery was well tolerated among the elderly.


Subject(s)
Genetic Vectors/administration & dosage , Macula Lutea/pathology , Vascular Endothelial Growth Factor Receptor-1/genetics , Visual Acuity , Wet Macular Degeneration/therapy , Aged , Aged, 80 and over , Female , Follow-Up Studies , Genetic Therapy/methods , Humans , Intravitreal Injections , Male , Middle Aged , Retrospective Studies , Time Factors , Tomography, Optical Coherence , Treatment Outcome , Wet Macular Degeneration/diagnosis
9.
Biomedicines ; 7(1)2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30871285

ABSTRACT

Healthy kidneys are important for the efficient regulation of metabolism. However, there is an ever increasing population of patients suffering from both acute and chronic kidney diseases that disrupt this homeostasis. This review will explore the emerging roles that interleukin 6 (IL-6) cytokine family members play in the pathogenesis of kidney disease. The IL-6 family of cytokines are involved in a diverse range of physiological functions. In relation to kidney disease, their involvement is no less diverse. Evidence from both preclinical and clinical sources show that IL-6 cytokine family members can play either a deleterious or protective role in response to kidney disease. This appears to be dependent on the type of kidney disease in question or the specific cytokine. Current attempts to use or target IL-6 cytokine family members as therapies of kidney diseases will be highlighted throughout this review. Finally, the involvement of IL-6 cytokine family members in kidney disease will be presented in the context of three regularly overlapping conditions: obesity, hypertension and diabetes.

10.
J Cell Physiol ; 233(1): 38-56, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28419469

ABSTRACT

The mechanisms responsible for the processing and quality control of the calcium-sensing receptor (CaSR) in the endoplasmic reticulum (ER) are largely unknown. In a yeast two-hybrid screen of the CaSR C-terminal tail (residues 865-1078), we identified osteosarcoma-9 (OS-9) protein as a binding partner. OS-9 is an ER-resident lectin that targets misfolded glycoproteins to the ER-associated degradation (ERAD) pathway through recognition of specific N-glycans by its mannose-6-phosphate receptor homology (MRH) domain. We show by confocal microscopy that the CaSR and OS-9 co-localize in the ER in COS-1 cells. In immunoprecipitation studies with co-expressed OS-9 and CaSR, OS-9 specifically bound the immature form of wild-type CaSR in the ER. OS-9 also bound the immature forms of a CaSR C-terminal deletion mutant and a C677A mutant that remains trapped in the ER, although binding to neither mutant was favored over wild-type receptor. OS-9 binding to immature CaSR required the MRH domain of OS-9 indicating that OS-9 acts as a lectin most likely to target misfolded CaSR to ERAD. Our results also identify two distinct binding interactions between OS-9 and the CaSR, one involving both C-terminal domains of the two proteins and the other involving both N-terminal domains. This suggests the possibility of more than one functional interaction between OS-9 and the CaSR. When we investigated the functional consequences of altered OS-9 expression, neither knockdown nor overexpression of OS-9 was found to have a significant effect on CaSR cell surface expression or CaSR-mediated ERK1/2 phosphorylation.


Subject(s)
Endoplasmic Reticulum/metabolism , Lectins/metabolism , Neoplasm Proteins/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum-Associated Degradation , Glycosylation , HEK293 Cells , Humans , Immunoprecipitation , Lectins/genetics , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Neoplasm Proteins/genetics , Phosphorylation , Protein Binding , Protein Folding , Protein Interaction Domains and Motifs , Proteolysis , RNA Interference , Receptors, Calcium-Sensing/genetics , Transfection , Two-Hybrid System Techniques
11.
Am J Ophthalmol ; 177: 150-158, 2017 May.
Article in English | MEDLINE | ID: mdl-28245970

ABSTRACT

PURPOSE: To assess the safety of rAAV.sFlt-1 subretinal injection in neovascular age-related macular degeneration (wet AMD) over 36 months. DESIGN: Phase 1 dose escalation trial. METHODS: Eight subjects with advanced, treatment-experienced wet AMD were randomly assigned (3:1) to treatment and non-gene therapy control groups. Eligible subjects were ≥65 years, had wet AMD, and had best-corrected visual acuity (BCVA) 10/200 to 20/80 in the study eye and 20/200 or better in the other eye. Three of the treatment group subjects received low-dose (1 × 1010 vector genomes) and 3 high-dose (1 × 1011 vector genomes) rAAV.sFLT-1 via subretinal injection. Study monitoring was monthly to the primary endpoint at month 12 and then protocol-driven follow-up study visits were conducted at months 18 and 36. All subjects received intravitreal ranibizumab at baseline and at week 4, and retreatment injections at subsequent visits based on prespecified criteria for active wet AMD. The primary endpoint was ocular and systemic safety, but exploratory data including BCVA, retinal center point thickness, and the number of ranibizumab retreatments at and between study visits were also analyzed. RESULTS: Six of the 8 subjects completed the 36-month study. Subretinal injection with pars plana vitrectomy was well tolerated in this cohort. No ocular or systemic safety signals were observed during the long-term follow-up period. Exploratory data analysis suggests stability of wet AMD over the 36-month period. CONCLUSIONS: Subretinal delivery of rAAV.sFLT-1 was well tolerated and demonstrated a favourable safety profile through month 36. Thus, rAAV.sFLT-1 could be safely considered for future evaluation in the treatment of wet AMD.


Subject(s)
Choroidal Neovascularization/therapy , Genetic Therapy/methods , Interleukin-1 Receptor-Like 1 Protein/administration & dosage , Visual Acuity , Wet Macular Degeneration/therapy , Aged , Aged, 80 and over , Choroidal Neovascularization/complications , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Injections , Male , Receptors, Interleukin-1 , Retina , Time Factors , Tomography, Optical Coherence , Treatment Outcome , Vitrectomy , Wet Macular Degeneration/physiopathology
12.
EBioMedicine ; 14: 168-175, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27865764

ABSTRACT

BACKGROUND: We present the results of a Phase 2a randomized controlled trial investigating the safety, and secondary endpoints of subretinal rAAV.sFLT-1 gene therapy in patients with active wet age-related macular degeneration (wAMD). METHODS: All patients (n=32), (ClinicalTrials.gov; NCT01494805), received ranibizumab injections at baseline and week 4, and thereafter according to prespecified criteria. Patients in the gene therapy group (n=21) received rAAV.sFLT-1 (1×1011vg). All patients were assessed every 4weeks to the week 52 primary endpoint. FINDINGS: Ocular adverse events (AEs) in the rAAV.sFLT-1 group were mainly procedure related and self-resolved. All 11 phakic patients in the rAAV.sFLT-1 group showed progression of cataract following vitrectomy. No systemic safety signals were observed and none of the serious AEs were associated with rAAV.sFLT-1. AAV2 capsid was not detected and rAAV.sFLT-1 DNA was detected transiently in the tears of 13 patients. ELISPOT analysis did not identify any notable changes in T-cell response. In the rAAV.sFLT-1 group 12 patients had neutralizing antibodies (nAb) to AAV2. There was no change in sFLT-1 levels in bodily fluids. In the rAAV.sFLT-1 group, Best Corrected Visual Acuity (BCVA) improved by a median of 1.0 (IQR: -3.0 to 9.0) Early Treatment Diabetic Retinopathy Study (ETDRS) letters from baseline compared to a median of -5.0 (IQR: -17.5 to 1.0) ETDRS letters change in the control group. Twelve (57%) patients in the rAAV.sFLT-1 group maintained or improved vision compared to 4 (36%) in the control group. The median number of ranibizumab retreatments was 2.0 (IQR: 1.0 to 6.0) for the gene therapy group compared to 4.0 (IQR: 3.5 to 4.0) for the control group. Interpretation rAAV.sFLT-1 combined with the option for co-treatment appears to be a safe and promising approach to the treatment of wAMD. FUNDING: National Health and Medical Research Council of Australia (AP1010405), Lions Eye Institute, Perth Australia, Avalanche Biotechnologies, Menlo Pk, CA, USA.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Wet Macular Degeneration/genetics , Wet Macular Degeneration/therapy , Aged , Aged, 80 and over , Case-Control Studies , Combined Modality Therapy , Dependovirus/immunology , Female , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/immunology , Humans , Male , Ranibizumab/administration & dosage , Ranibizumab/therapeutic use , Retina/metabolism , Retina/pathology , Tissue Distribution , Tomography, Optical Coherence , Treatment Outcome , Wet Macular Degeneration/diagnosis , Wet Macular Degeneration/drug therapy
13.
Lancet ; 386(10011): 2395-403, 2015 Dec 12.
Article in English | MEDLINE | ID: mdl-26431823

ABSTRACT

BACKGROUND: Neovascular, or wet, age-related macular degeneration causes central vision loss and represents a major health problem in elderly people, and is currently treated with frequent intraocular injections of anti-VEGF protein. Gene therapy might enable long-term anti-VEGF therapy from a single treatment. We tested the safety of rAAV.sFLT-1 in treatment of wet age-related macular degeneration with a single subretinal injection. METHODS: In this single-centre, phase 1, randomised controlled trial, we enrolled patients with wet age-related macular degeneration at the Lions Eye Institute and the Sir Charles Gairdner Hospital (Nedlands, WA, Australia). Eligible patients had to be aged 65 years or older, have age-related macular degeneration secondary to active subfoveal choroidal neovascularisation, with best corrected visual acuity (BCVA) of 3/60-6/24 and 6/60 or better in the other eye. Patients were randomly assigned (3:1) to receive either 1 × 10(10) vector genomes (vg; low-dose rAAV.sFLT-1 group) or 1 × 10(11) vg (high-dose rAAV.sFLT-1 group), or no gene-therapy treatment (control group). Randomisation was done by sequential group assignment. All patients and investigators were unmasked. Staff doing the assessments were masked to the study group at study visits. All patients received ranibizumab at baseline and week 4, and rescue treatment during follow-up based on prespecified criteria including BCVA measured on the Early Treatment Diabetic Retinopathy Study (EDTRS) scale, optical coherence tomography, and fluorescein angiography. The primary endpoint was ocular and systemic safety. This trial is registered with ClinicalTrials.gov, number NCT01494805. FINDINGS: From Dec 16, 2011, to April 5, 2012, we enrolled nine patients of whom eight were randomly assigned to receive either intervention (three patients in the low-dose rAAV.sFLT-1 group and three patients in the high-dose rAAV.sFLT-1 group) or no treatment (two patients in the control group). Subretinal injection of rAAV.sFLT-1 was highly reproducible. No drug-related adverse events were noted; procedure-related adverse events (subconjunctival or subretinal haemorrhage and mild cell debris in the anterior vitreous) were generally mild and self-resolving. There was no evidence of chorioretinal atrophy. Clinical laboratory assessments generally remained unchanged from baseline. Four (67%) of six patients in the treatment group required zero rescue injections, and the other two (33%) required only one rescue injection each. INTERPRETATION: rAAV.sFLT-1 was safe and well tolerated. These results support ocular gene therapy as a potential long-term treatment option for wet age-related macular degeneration. FUNDING: National Health and Medical Research Council of Australia, Richard Pearce Bequest, Lions Save Sight Foundation, Brian King Fellowship, and Avalanche Biotechnologies, Inc.


Subject(s)
Genetic Therapy/methods , Vascular Endothelial Growth Factor Receptor-1/administration & dosage , Wet Macular Degeneration/therapy , Adenoviridae , Aged , Aged, 80 and over , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Choroidal Neovascularization/complications , Choroidal Neovascularization/physiopathology , Choroidal Neovascularization/therapy , Female , Genetic Therapy/adverse effects , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Humans , Injections, Intraocular , Male , Ranibizumab/administration & dosage , Ranibizumab/adverse effects , Recombinant Proteins , Vascular Endothelial Growth Factor Receptor-1/adverse effects , Visual Acuity , Wet Macular Degeneration/etiology , Wet Macular Degeneration/physiopathology
14.
Exp Eye Res ; 122: 123-31, 2014 May.
Article in English | MEDLINE | ID: mdl-24703908

ABSTRACT

The molecular mechanisms of vascular leakage in diabetic macular edema and proliferative retinopathy are poorly understood, mainly due to the lack of reliable in vivo models. The Akimba (Ins2(Akita)VEGF(+/-)) mouse model combines retinal neovascularization with hyperglycemia, and in contrast to other models, displays the majority of signs of advanced clinical diabetic retinopathy (DR). To study the molecular mechanism that underlies the breakdown of the blood-retinal barrier (BRB) in diabetic macular edema and proliferative diabetic retinopathy, we investigated the retinal vasculature of Akimba and its parental mice Kimba (trVEGF029) and Akita (Ins2(Akita)). Quantitative PCR, immunohistochemistry and fluorescein angiography were used to characterize the retinal vasculature with special reference to the inner BRB. Correlations between the degree of fluorescein leakage and retinal gene expression were tested by calculating the Spearman's correlation coefficient. Fluorescein leakage demonstrating BRB loss was observed in Kimba and Akimba, but not in Akita or wild type mice. In Kimba and Akimba mice fluorescein leakage was associated with focal angiogenesis and correlated significantly with Plvap gene expression. PLVAP is an endothelial cell-specific protein that is absent in intact blood-retinal barrier, but its expression significantly increases in pathological conditions such as DR. Furthermore, in Akimba mice BRB disruption was linked to decreased expression of endothelial junction proteins, pericyte dropout and vessel loss. Despite fluorescein leakage, no alteration in BRB protein levels or pericyte coverage was detected in retinas of Kimba mice. In summary, our data not only demonstrate that hyperglycemia sensitizes retinal vasculature to the effects of VEGF, leading to more severe microvascular changes, but also confirm an important role of PLVAP in the regulation of BRB permeability.


Subject(s)
Blood-Retinal Barrier/pathology , Diabetic Retinopathy/genetics , Disease Models, Animal , Retinal Neovascularization/genetics , Retinal Vessels/pathology , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Capillary Permeability , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chemokines/genetics , Chemokines/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Endoglin , Fluorescein Angiography , Gene Expression , Hyperglycemia/genetics , Hyperglycemia/pathology , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Macular Edema/genetics , Macular Edema/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Pericytes/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Real-Time Polymerase Chain Reaction , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
15.
Clin Biochem ; 45(12): 943-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22503956

ABSTRACT

Following the discovery of the calcium-sensing receptor (CaSR) in 1993, its pivotal role in disorders of calcium homeostasis such as Familial Hypocalciuric Hypercalcemia (FHH) was quickly demonstrated. Since then, it has become clear that the CaSR has immense functional versatility largely through its ability to activate many different signaling pathways in a ligand- and tissue-specific manner. This allows the receptor to play diverse and crucial roles in human physiology and pathophysiology, both in calcium homeostasis and in tissues and biological processes unrelated to calcium balance. This review covers current knowledge of the role of the CaSR in disorders of calcium homeostasis (FHH, neonatal severe hyperparathyroidism, autosomal dominant hypocalcemia, primary and secondary hyperparathyroidism, hypercalcemia of malignancy) as well as unrelated diseases such as breast and colorectal cancer (where the receptor appears to play a tumor suppressor role), Alzheimer's disease, pancreatitis, diabetes mellitus, hypertension and bone and gastrointestinal disorders. In addition, it examines the use or potential use of CaSR agonists or antagonists (calcimimetics and calcilytics) and other drugs mediated through the CaSR, in the management of disorders as diverse as hyperparathyroidism, osteoporosis and gastrointestinal disease.


Subject(s)
Receptors, Calcium-Sensing/physiology , Animals , Bone Diseases/drug therapy , Bone Diseases/metabolism , Brain Diseases/metabolism , Calcium Metabolism Disorders/etiology , Calcium Metabolism Disorders/metabolism , Cardiovascular Diseases/metabolism , Gastrointestinal Diseases/metabolism , Humans , Hyperparathyroidism/metabolism , Mutation , Neoplasms/complications , Neoplasms/metabolism , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism
16.
Biochem J ; 441(3): 995-1006, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22010828

ABSTRACT

A yeast two-hybrid screen performed to identify binding partners of the CaR (calcium-sensing receptor) intracellular tail identified the adaptor protein 14-3-3θ as a novel binding partner that bound to the proximal membrane region important for CaR expression and signalling. The 14-3-3θ protein directly interacted with the CaR tail in pull-down studies and FLAG-tagged CaR co-immunoprecipitated with EGFP (enhanced green fluorescent protein)-tagged 14-3-3θ when co-expressed in HEK (human embryonic kidney)-293 or COS-1 cells. The interaction between the CaR and 14-3-3θ did not require a putative binding site in the membrane-proximal region of the CaR tail and was independent of PKC (protein kinase C) phosphorylation. Confocal microscopy demonstrated co-localization of the CaR and EGFP-14-3-3θ in the ER (endoplasmic reticulum) of HEK-293 cells that stably expressed the CaR (HEK-293/CaR cells), but 14-3-3θ overexpression had no effect on membrane expression of the CaR. Overexpression of 14-3-3θ in HEK-293/CaR cells attenuated CaR-mediated Rho signalling, but had no effect on ERK (extracellular-signal-regulated kinase) 1/2 signalling. Another isoform identified from the library, 14-3-3ζ, exhibited similar behaviour to that of 14-3-3θ with respect to CaR tail binding, cellular co-localization and impact on receptor-mediated signalling. However, unlike 14-3-3θ, this isoform, when overexpressed, significantly reduced CaR plasma membrane expression. Results indicate that 14-3-3 proteins mediate CaR-dependent Rho signalling and may modulate the plasma membrane expression of the CaR.


Subject(s)
14-3-3 Proteins/metabolism , Receptors, Calcium-Sensing/metabolism , rho-Associated Kinases/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
17.
Biochem Biophys Res Commun ; 412(4): 584-9, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21843504

ABSTRACT

The calcium-sensing receptor (CaR) plays an integral role in calcium homeostasis and the regulation of other cellular functions including cell proliferation and cytoskeletal organisation. The multifunctional nature of the CaR is manifested through ligand-dependent stimulation of different signalling pathways that are also regulated by partner binding proteins. Following a yeast two-hybrid library screen using the intracellular tail of the CaR as bait, we identified several novel binding partners including the focal adhesion protein, testin. Testin has not previously been shown to interact with cell surface receptors. The sites of interaction between the CaR and testin were mapped to the membrane proximal region of the receptor tail and the second zinc-finger of LIM domain 1 of testin, the integrity of which was found to be critical for the CaR-testin interaction. The CaR-testin association was confirmed in HEK293 cells by coimmunoprecipitation and confocal microscopy studies. Ectopic expression of testin in HEK293 cells stably expressing the CaR enhanced CaR-stimulated Rho activity but had no effect on CaR-stimulated ERK signalling. These results suggest an interplay between the CaR and testin in the regulation of CaR-mediated Rho signalling with possible effects on the cytoskeleton.


Subject(s)
Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Receptors, Calcium-Sensing/metabolism , rho-Associated Kinases/metabolism , Amino Acid Sequence , Animals , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Immunoprecipitation , LIM Domain Proteins/genetics , Mice , Molecular Sequence Data , Protein Structure, Tertiary , RNA-Binding Proteins , Receptors, Calcium-Sensing/genetics , Signal Transduction , Two-Hybrid System Techniques
18.
Endocr Rev ; 32(1): 3-30, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20729338

ABSTRACT

Compelling evidence of a cell surface receptor sensitive to extracellular calcium was observed as early as the 1980s and was finally realized in 1993 when the calcium-sensing receptor (CaR) was cloned from bovine parathyroid tissue. Initial studies relating to the CaR focused on its key role in extracellular calcium homeostasis, but as the amount of information about the receptor grew it became evident that it was involved in many biological processes unrelated to calcium homeostasis. The CaR responds to a diverse array of stimuli extending well beyond that merely of calcium, and these stimuli can lead to the initiation of a wide variety of intracellular signaling pathways that in turn are able to regulate a diverse range of biological processes. It has been through the examination of the molecular characteristics of the CaR that we now have an understanding of how this single receptor is able to convert extracellular messages into specific cellular responses. Recent CaR-related reviews have focused on specific aspects of the receptor, generally in the context of the CaR's role in physiology and pathophysiology. This review will provide a comprehensive exploration of the different aspects of the receptor, including its structure, stimuli, signalling, interacting protein partners, and tissue expression patterns, and will relate their impact on the functionality of the CaR from a molecular perspective.


Subject(s)
Receptors, Calcium-Sensing/metabolism , Animals , Bone and Bones/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Cattle , Gastrointestinal Tract/metabolism , Homeostasis/physiology , Humans , Kidney/metabolism , Mice , Parathyroid Glands/metabolism , Rats , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/physiology , Signal Transduction
19.
J Bone Miner Res ; 24(7): 1216-23, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19257822

ABSTRACT

Previously reported Sequestosome 1(SQSTM1)/p62 gene mutations associated with Paget's disease of bone (PDB) cluster in, or cause deletion of, the ubiquitin-associated (UBA) domain. The aims of this study were to examine the prevalence of SQSTM1 mutations in Australian patients, genotype/phenotype correlations and the functional consequences of a novel point mutation (P364S) located upstream of the UBA. Mutation screening of the SQSTM1 gene was conducted on 49 kindreds with PDB. In addition, 194 subjects with apparently sporadic PDB were screened for the common P392L mutation by restriction enzyme digestion. HEK293 cells stably expressing RANK were co-transfected with expression plasmids for SQSTM1 (wildtype or mutant) or empty vector and a NF-kappaB luciferase reporter gene. GST-SQSTM1 (wildtype and mutant) proteins were used in pull-down assays to compare monoubiquitin-binding ability. We identified SQSTM1 mutations in 12 of 49 families screened (24.5%), comprising 9 families with the P392L mutation and 1 family each with the following mutations: K378X, 390X, and a novel P364S mutation in exon 7, upstream of the UBA. The P392L mutation was found in 9 of 194 (4.6%) patients with sporadic disease. Subjects with SQSTM1 mutations had more extensive disease, but not earlier onset, compared with subjects without mutations. In functional studies, the P364S mutation increased NF-kappaB activation compared with wildtype SQSTM1 but did not reduce ubiquitin binding. This suggests that increased NF-kappaB signaling, but not the impairment of ubiquitin binding, may be essential in the pathogenesis of PDB associated with SQSTM1 mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Mutation, Missense , NF-kappa B/metabolism , Osteitis Deformans/genetics , Osteitis Deformans/metabolism , Signal Transduction , Ubiquitin/metabolism , Adult , Aged , Amino Acid Substitution , Australia/epidemiology , Cell Line , Female , Genotype , Humans , Male , Middle Aged , NF-kappa B/genetics , Osteitis Deformans/epidemiology , Pedigree , Phenotype , Prevalence , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Sequestosome-1 Protein
20.
Clin Endocrinol (Oxf) ; 64(5): 580-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16649980

ABSTRACT

OBJECTIVE: Heterozygous inactivating mutations of the calcium-sensing receptor (CaR) gene cause familial hypocalciuric hypercalcaemia (FHH), a generally benign disorder characterized by mild to moderate PTH-dependent hypercalcaemia. We aimed to identify the causative CaR mutations in three families with FHH and examine the correlation between type of mutation and biochemical and/or functional phenotypes. PATIENTS, DESIGN AND MEASUREMENTS: The CaR gene from FHH family members was assessed for mutations by direct DNA sequencing and mutations were confirmed by restriction enzyme analysis. Functional studies on two missense mutations were conducted by introducing them by site-directed mutagenesis into the CaR cloned into a mammalian expression vector, and assessing calcium responsiveness using an inositol phosphate (IP) assay in HEK293 cells. Biochemical data from patients heterozygous for each type of mutant were correlated with functionality. RESULTS: Two novel nonsense mutations (R25stop and K323stop) and one novel missense mutation (G778D) were identified. The G778D mutant receptor and another mutation identified in an earlier study (L174R) demonstrated a complete lack of Ca2+ responsiveness using the IP assay. When cotransfected with wild-type receptor, the mutant receptors demonstrated a dominant-negative effect on wild-type receptor response, with L174R having a more pronounced effect than G778D. Significantly more severe hypercalcaemia and a trend towards higher PTH levels were observed in patients heterozygous for CaR mutants with a stronger dominant-negative effect. CONCLUSIONS: Naturally occurring CaR mutations with differences in dominant-negative effect on wild-type receptor demonstrate differences in biochemical severity in FHH.


Subject(s)
Calcium/urine , Hypercalcemia/genetics , Mutation, Missense , Receptors, Calcium-Sensing/genetics , Animals , Biological Assay , Calcium/metabolism , Calcium/pharmacology , Cell Line , DNA Mutational Analysis , Heterozygote , Humans , Hypercalcemia/metabolism , Least-Squares Analysis , Mutagenesis, Site-Directed , Receptors, Calcium-Sensing/analysis , Receptors, Calcium-Sensing/metabolism , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...