Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Am J Pathol ; 194(6): 1090-1105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38403162

ABSTRACT

Changes in the anterior segment of the eye due to type 2 diabetes mellitus (T2DM) are not well-characterized, in part due to the lack of a reliable animal model. This study evaluated changes in the anterior segment, including crystalline lens health, corneal endothelial cell density, aqueous humor metabolites, and ciliary body vasculature, in a rat model of T2DM compared with human eyes. Male Sprague-Dawley rats were fed a high-fat diet (45% fat) or normal diet, and rats fed the high-fat diet were injected with streptozotocin intraperitoneally to generate a model of T2DM. Cataract formation and corneal endothelial cell density were assessed using microscopic analysis. Diabetes-related rat aqueous humor alterations were assessed using metabolomics screening. Transmission electron microscopy was used to assess qualitative ultrastructural changes ciliary process microvessels at the site of aqueous formation in the eyes of diabetic rats and humans. Eyes from the diabetic rats demonstrated cataracts, lower corneal endothelial cell densities, altered aqueous metabolites, and ciliary body ultrastructural changes, including vascular endothelial cell activation, pericyte degeneration, perivascular edema, and basement membrane reduplication. These findings recapitulated diabetic changes in human eyes. These results support the use of this model for studying ocular manifestations of T2DM and support a hypothesis postulating blood-aqueous barrier breakdown and vascular leakage at the ciliary body as a mechanism for diabetic anterior segment pathology.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Rats, Sprague-Dawley , Animals , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Male , Rats , Humans , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Anterior Eye Segment/pathology , Aqueous Humor/metabolism , Cataract/pathology , Cataract/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/metabolism , Lens, Crystalline/ultrastructure , Ciliary Body/pathology , Ciliary Body/metabolism , Diet, High-Fat/adverse effects
2.
PLoS One ; 18(1): e0280491, 2023.
Article in English | MEDLINE | ID: mdl-36652491

ABSTRACT

Diabetes mellitus is a multiorgan systemic disease impacting numerous ocular structures that results in significant ocular morbidity and often results in more frequent corneal and glaucoma surgeries for affected individuals. We hypothesize that the systemic metabolic and proteomic derangement observed in the progression of diabetes influences the composition of the aqueous humor (AH), which ultimately impacts the anterior segment health of the eye. To identify changes associated with diabetes progression, we mapped the metabolite profile and proteome of AH samples from patients with varying severities of type II diabetes (T2DM). Patients were classified as nondiabetic (ND or control), non-insulin-dependent diabetic without advanced features of disease (NAD-ni), insulin-dependent diabetic without advanced features (NAD-i), or diabetic with advanced features (AD). AH samples collected from the anterior chamber during elective ophthalmic surgery were evaluated for metabolite and protein expression changes associated with diabetic severity via gas chromatography/mass spectrometry and ultra-high performance liquid chromatography tandem mass spectrometry, respectively. Metabolic and proteomic pathway analyses were conducted utilizing MetaboAnalyst 4.0 and Ingenuity Pathway Analysis. A total of 14 control, 12 NAD-ni, 4 NAD-I, and 14 AD samples were included for analysis. Elevated levels of several branched amino acids (e.g., valine, leucine, isoleucine), and lipid metabolites (e.g., palmitate) were found only with increasing diabetic severity (i.e., the AD group). Similar proteomic trends were noted in amino acid and fatty acid metabolism and the unfolded protein/stress response. These results represent the first report of both metabolomic and proteomic evaluation of aqueous humor. Diabetes results in metabolic and proteomic perturbations detectable in the AH, and unique changes become manifest as T2DM severity worsens. Changes in AH composition may serve as an indicator of disease severity, risk assessment of anterior segment cells and structures, and potential future therapies.


Subject(s)
Aqueous Humor , Diabetes Mellitus, Type 2 , Humans , Aqueous Humor/metabolism , Diabetes Mellitus, Type 2/metabolism , Proteomics , NAD/metabolism , Chromatography, Liquid
3.
Biomaterials ; 275: 120842, 2021 08.
Article in English | MEDLINE | ID: mdl-34087583

ABSTRACT

Defective cellular metabolism, impaired mitochondrial function, and increased cell death are major problems that adversely affect donor tissues during hypothermic preservation prior to transplantation. These problems are thought to arise from accumulated reactive oxygen species (ROS) inside cells. Oxidative stress acting on the cells of organs and tissues preserved in hypothermic conditions before surgery, as is the case for cornea transplantation, is thought to be a major reason behind cell death prior to surgery and decreased graft survival after transplantation. We have recently discovered that ubiquinol - the reduced and active form of coenzyme Q10 and a powerful antioxidant - significantly enhances mitochondrial function and reduces apoptosis in human donor corneal endothelial cells. However, ubiquinol is highly lipophilic, underscoring the need for an aqueous-based formulation of this molecule. Herein, we report a highly dispersible and stable formulation comprising a complex of ubiquinol and gamma cyclodextrin (γ-CD) for use in aqueous-phase ophthalmic products. Docking studies showed that γ-CD has the strongest binding affinity with ubiquinol compared to α- or ß-CD. Complexed ubiquinol showed significantly higher stability compared to free ubiquinol in different aqueous ophthalmic products including Optisol-GS® corneal storage medium, balanced salt solution for intraocular irrigation, and topical Refresh® artificial tear eye drops. Greater ROS scavenging activity was noted in a cell model with high basal metabolism and ROS generation (A549) and in HCEC-B4G12 human corneal endothelial cells after treatment with ubiquinol/γ-CD compared to free ubiquinol. Furthermore, complexed ubiquinol was more effective at lowering ROS, and at far lower concentrations, compared to free ubiquinol. Complexed ubiquinol inhibited lipid peroxidation and protected HCEC-B4G12 cells against erastin-induced ferroptosis. No evidence of cellular toxicity was detected in HCEC-B4G12 cells after treatment with complexed ubiquinol. Using a vertical diffusion system, a topically applied inclusion complex of γ-CD and a lipophilic dye (coumarin-6) demonstrated transcorneal penetrance in porcine corneas and the capacity for the γ-CD vehicle to deliver drug to the corneal endothelium. Using the same model, topically applied ubiquinol/γ-CD complex penetrated the entire thickness of human donor corneas with markedly greater ubiquinol retention in the endothelium compared to free ubiquinol. Lastly, the penetrance of ubiquinol/γ-CD complex was assayed using human donor corneas preserved for 7 days in Optisol-GS® per standard industry practices, and demonstrated higher amounts of ubiquinol retained in the corneal endothelium compared to free ubiquinol. In summary, ubiquinol complexed with γ-CD is a highly stable composition that can be incorporated into a variety of aqueous-phase products for ophthalmic use including donor corneal storage media and topical eye drops to scavenge ROS and protect corneal endothelial cells against oxidative damage.


Subject(s)
Corneal Transplantation , Endothelial Cells , Animals , Cornea , Culture Media, Serum-Free , Dextrans , Endothelium, Corneal , Gentamicins , Humans , Organ Preservation , Swine , Ubiquinone/analogs & derivatives
4.
Invest Ophthalmol Vis Sci ; 62(3): 22, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33724294

ABSTRACT

Mitochondrial function is essential for the viability of aerobic eukaryotic cells, as mitochondria provide energy through the generation of adenosine triphosphate (ATP), regulate cellular metabolism, provide redox balancing, participate in immune signaling, and can initiate apoptosis. Mitochondria are dynamic organelles that participate in a cyclical and ongoing process of regeneration and autophagy (clearance), termed mitophagy specifically for mitochondrial (macro)autophagy. An imbalance in mitochondrial function toward mitochondrial dysfunction can be catastrophic for cells and has been characterized in several common ophthalmic diseases. In this article, we review mitochondrial homeostasis in detail, focusing on the balance of mitochondrial dynamics including the processes of fission and fusion, and provide a description of the mechanisms involved in mitophagy. Furthermore, this article reviews investigations of ocular diseases with impaired mitophagy, including Fuchs endothelial corneal dystrophy, primary open-angle glaucoma, diabetic retinopathy, and age-related macular degeneration, as well as several primary mitochondrial diseases with ocular phenotypes that display impaired mitophagy, including mitochondrial encephalopathy lactic acidosis stroke, Leber hereditary optic neuropathy, and chronic progressive external ophthalmoplegia. The results of various studies using cell culture, animal, and human tissue models are presented and reflect a growing awareness of mitophagy impairment as an important feature of ophthalmic disease pathology. As this review indicates, it is imperative that mitophagy be investigated as a targetable mechanism in developing therapies for ocular diseases characterized by oxidative stress and mitochondrial dysfunction.


Subject(s)
Diabetic Retinopathy/physiopathology , Fuchs' Endothelial Dystrophy/physiopathology , Glaucoma, Open-Angle/physiopathology , Macular Degeneration/physiopathology , Mitochondria/physiology , Mitochondrial Diseases/physiopathology , Mitophagy/physiology , Animals , Humans , Molecular Targeted Therapy
5.
Cornea ; 39(10): 1285-1290, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32558735

ABSTRACT

PURPOSE: To determine whether ubiquinol improves mitochondrial function and cell viability in human donor corneal endothelial cells during hypothermic corneal tissue storage. METHODS: Endothelial cell Descemet membrane tissues were treated with 10 µM ubiquinol, the reduced form of the antioxidant coenzyme Q10, for 5 days in Optisol-GS storage media before assaying for mitochondrial activity using extracellular flux analysis of oxygen consumption. In addition, endothelial cell Descemet membrane tissues were analyzed for cell viability using apoptosis and necrosis assays. Control tissues from mate corneas were treated with diluent only, and comparisons were analyzed for differences. RESULTS: A total of 13 donor corneal tissues with a mean (SEM) preservation time of 11.8 days (0.4) were included for the analysis. Treatment with 10 µM ubiquinol increased spare respiratory capacity by 174% (P = 0.001), maximal respiration by 93% (P = 0.003), and proton leak by 80% (P = 0.047) compared with controls. Cells treated with ubiquinol had no significant change in cell necrosis or apoptosis. CONCLUSIONS: Preliminary testing in donor corneal tissue at specified doses indicates that ubiquinol may be a useful biocompatible additive to hypothermic corneal storage media that increases corneal endothelial cell mitochondrial function. Additional investigations are indicated to further study and optimize the dose and formulation of ubiquinol for use in preserving donor corneal tissue function during hypothermic storage.


Subject(s)
Cell Respiration/physiology , Endothelium, Corneal/drug effects , Micronutrients/pharmacology , Mitochondria/metabolism , Ubiquinone/analogs & derivatives , Aged , Cell Count , Cell Survival/drug effects , Chondroitin Sulfates , Complex Mixtures , Cryopreservation , Descemet Membrane/drug effects , Dextrans , Female , Gentamicins , Humans , Male , Middle Aged , Organ Preservation , Organ Preservation Solutions , Tissue Donors , Ubiquinone/pharmacology
6.
J Cell Sci ; 132(5)2019 02 15.
Article in English | MEDLINE | ID: mdl-30665891

ABSTRACT

Bardet-Beidl syndrome (BBS) manifests from genetic mutations encoding for one or more BBS proteins. BBS4 loss impacts olfactory ciliation and odor detection, yet the cellular mechanisms remain unclear. Here, we report that Bbs4-/- mice exhibit shorter and fewer olfactory sensory neuron (OSN) cilia despite retaining odorant receptor localization. Within Bbs4-/- OSN cilia, we observed asynchronous rates of IFT-A/B particle movements, indicating miscoordination in IFT complex trafficking. Within the OSN dendritic knob, the basal bodies are dynamic, with incorporation of ectopically expressed centrin-2 and γ-tubulin occurring after nascent ciliogenesis. Importantly, BBS4 loss results in the reduction of basal body numbers separate from cilia loss. Adenoviral expression of BBS4 restored OSN cilia lengths and was sufficient to re-establish odor detection, but failed to rescue ciliary and basal body numbers. Our results yield a model for the plurality of BBS4 functions in OSNs that includes intraciliary and periciliary roles that can explain the loss of cilia and penetrance of ciliopathy phenotypes in olfactory neurons.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Cilia/physiology , Flagella/metabolism , Microtubule-Associated Proteins/metabolism , Olfactory Receptor Neurons/physiology , Animals , Basal Bodies/pathology , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Phenotype , Protein Transport , Smell , Trimethoprim, Sulfamethoxazole Drug Combination/metabolism , Tubulin/metabolism
7.
PLoS Genet ; 13(10): e1007057, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29049287

ABSTRACT

Genetic mutations disrupting the structure and function of primary cilia cause various inherited retinal diseases in humans. Bardet-Biedl syndrome (BBS) is a genetically heterogeneous, pleiotropic ciliopathy characterized by retinal degeneration, obesity, postaxial polydactyly, intellectual disability, and genital and renal abnormalities. To gain insight into the mechanisms of retinal degeneration in BBS, we developed a congenital knockout mouse of Bbs8, as well as conditional mouse models in which function of the BBSome (a protein complex that mediates ciliary trafficking) can be temporally inactivated or restored. We demonstrate that BBS mutant mice have defects in retinal outer segment morphogenesis. We further demonstrate that removal of Bbs8 in adult mice affects photoreceptor function and disrupts the structural integrity of the outer segment. Notably, using a mouse model in which a gene trap inhibiting Bbs8 gene expression can be removed by an inducible FLP recombinase, we show that when BBS8 is restored in immature retinas with malformed outer segments, outer segment extension can resume normally and malformed outer segment discs are displaced distally by normal outer segment structures. Over time, the retinas of the rescued mice become morphologically and functionally normal, indicating that there is a window of plasticity when initial retinal outer segment morphogenesis defects can be ameliorated.


Subject(s)
Morphogenesis/physiology , Photoreceptor Cells/metabolism , Protein Transport/physiology , Animals , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/metabolism , Bardet-Biedl Syndrome/pathology , Cilia/metabolism , Mice , Mice, Knockout , Models, Animal , Morphogenesis/genetics , Mutation/genetics , Protein Transport/genetics , Retina/metabolism , Retina/physiology
8.
Mol Ther ; 25(4): 904-916, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28237838

ABSTRACT

Olfactory dysfunction is a pervasive but underappreciated health concern that affects personal safety and quality of life. Patients with olfactory dysfunctions have limited therapeutic options, particularly those involving congenital diseases. Bardet-Biedl syndrome (BBS) is one such disorder, where olfactory loss and other symptoms manifest from defective cilium morphology and/or function in various cell types/tissues. Olfactory sensory neurons (OSNs) of BBS mutant mice lack the capacity to build/maintain cilia, rendering the cells incapable of odor detection. Here we examined OSN cilium defects in Bbs1 mutant mice and assessed the utility of gene therapy to restore ciliation and function in young and adult mice. Bbs1 mutant mice possessed short residual OSN cilia in which BBSome protein trafficking and odorant detection were defective. Gene therapy with an adenovirus-delivered wild-type Bbs1 gene restored OSN ciliation, corrected BBSome cilium trafficking defects, and returned acute odor responses. Finally, using clinically approved AAV serotypes, we demonstrate, for the first time, the capacity of AAVs to restore ciliation and odor detection in OSNs of Bbs1 mutants. Together, our data demonstrate that OSN ciliogenesis can be promoted in differentiated cells of young and adult Bbs1 mutants and highlight the potential of gene therapy as a viable restorative treatment for congenital olfactory disorders.


Subject(s)
Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/physiopathology , Genetic Therapy , Olfactory Receptor Neurons/metabolism , Alleles , Animals , Bardet-Biedl Syndrome/therapy , Cilia/metabolism , Cilia/pathology , Dependovirus/genetics , Disease Models, Animal , Ectopic Gene Expression , Gene Expression , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Olfactory Perception/genetics , Phenotype , Protein Transport , Transduction, Genetic
9.
PLoS Genet ; 12(2): e1005890, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26926121

ABSTRACT

Bardet-Biedl syndrome (BBS) is a highly pleiotropic autosomal recessive disorder associated with a wide range of phenotypes including obesity. However, the underlying mechanism remains unclear. Here, we show that neuronal BBSome is a critical determinant of energy balance through its role in the regulation of the trafficking of the long signaling form of the leptin receptor (LRb). Targeted disruption of the BBSome by deleting the Bbs1 gene from the nervous system causes obesity in mice, and this phenotype is reproduced by ablation of the Bbs1 gene selectively in the LRb-expressing cells, but not from adipocytes. Obesity developed as a consequence of both increased food intake and decreased energy expenditure in mice lacking the Bbs1 gene in LRb-expressing cells. Strikingly, the well-known role of BBS proteins in the regulation of ciliary formation and function is unlikely to account for the obesogenic effect of BBS1 loss as disruption of the intraflagellar transport (IFT) machinery required for ciliogenesis by deleting the Ift88 gene in LRb-expressing cells caused a marginal increase in body weight and adiposity. Instead, we demonstrate that silencing BBS proteins, but not IFT88, impair the trafficking of the LRb to the plasma membrane leading to central leptin resistance in a manner independent of obesity. Our data also demonstrate that postnatal deletion of the Bbs1 gene in the mediobasal hypothalamus can cause obesity in mice, arguing against an early neurodevelopmental origin of obesity in BBS. Our results depict a novel mechanism underlying energy imbalance and obesity in BBS with potential implications in common forms of human obesity.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Cell Membrane/metabolism , Receptors, Leptin/metabolism , Animals , Bardet-Biedl Syndrome/genetics , Cell Membrane/genetics , Energy Metabolism/physiology , Female , Hypothalamus/physiology , Mice, Mutant Strains , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/metabolism , Obesity/genetics , Obesity/metabolism , Protein Transport , Receptors, Leptin/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
J Cell Sci ; 126(Pt 11): 2372-80, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23572516

ABSTRACT

Bardet-Biedl Syndrome (BBS) is a pleiotropic and genetically heterozygous disorder caused independently by numerous genes (BBS1-BBS17). Seven highly conserved BBS proteins (BBS1, 2, 4, 5, 7, 8 and 9) form a complex known as the BBSome, which functions in ciliary membrane biogenesis. BBS7 is both a unique subunit of the BBSome and displays direct physical interaction with a second BBS complex, the BBS chaperonin complex. To examine the in vivo function of BBS7, we generated Bbs7 knockout mice. Bbs7(-/-) mice show similar phenotypes to other BBS gene mutant mice including retinal degeneration, obesity, ventriculomegaly and male infertility characterized by abnormal spermatozoa flagellar axonemes. Using tissues from Bbs7(-/-) mice, we show that BBS7 is required for BBSome formation, and that BBS7 and BBS2 depend on each other for protein stability. Although the BBSome serves as a coat complex for ciliary membrane proteins, BBS7 is not required for the localization of ciliary membrane proteins polycystin-1, polycystin-2, or bitter taste receptors, but absence of BBS7 leads to abnormal accumulation of the dopamine D1 receptor to the ciliary membrane, indicating that BBS7 is involved in specific membrane protein localization to cilia.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Carrier Proteins/metabolism , Cell Membrane/metabolism , Molecular Chaperones/metabolism , Multiprotein Complexes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/pathology , Carrier Proteins/genetics , Cell Membrane/genetics , Cell Membrane/pathology , Cilia/genetics , Cilia/metabolism , Cilia/pathology , Cytoskeletal Proteins , Disease Models, Animal , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Mice, Knockout , Molecular Chaperones/genetics , Multiprotein Complexes/genetics , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
12.
Nat Med ; 18(12): 1797-804, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160237

ABSTRACT

Hydrocephalus is a common neurological disorder that leads to expansion of the cerebral ventricles and is associated with a high rate of morbidity and mortality. Most neonatal cases are of unknown etiology and are likely to have complex inheritance involving multiple genes and environmental factors. Identifying molecular mechanisms for neonatal hydrocephalus and developing noninvasive treatment modalities are high priorities. Here we use a hydrocephalic mouse model of the human ciliopathy Bardet-Biedl Syndrome (BBS) and identify a role for neural progenitors in the pathogenesis of neonatal hydrocephalus. We found that hydrocephalus in this mouse model is caused by aberrant platelet-derived growth factor receptor α (PDGFR-α) signaling, resulting in increased apoptosis and impaired proliferation of chondroitin sulfate proteoglycan 4 (also known as neuron-glial antigen 2 or NG2)(+)PDGFR-α(+) neural progenitors. Targeting this pathway with lithium treatment rescued NG2(+)PDGFR-α(+) progenitor cell proliferation in BBS mutant mice, reducing their ventricular volume. Our findings demonstrate that neural progenitors are crucial in the pathogenesis of neonatal hydrocephalus, and we identify new therapeutic targets for this common neurological disorder.


Subject(s)
Antigens/metabolism , Apoptosis/physiology , Bardet-Biedl Syndrome/pathology , Hydrocephalus/etiology , Neural Stem Cells/cytology , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Bromodeoxyuridine , Cell Proliferation/drug effects , DNA Primers/genetics , Female , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Lithium/pharmacology , Magnetic Resonance Imaging , Male , Mice , Mice, Mutant Strains , Neural Stem Cells/metabolism , Real-Time Polymerase Chain Reaction
13.
Vision Res ; 75: 77-87, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22940089

ABSTRACT

PURPOSE: To characterize the phenotype of Bardet-Biedl syndrome (BBS) patients homozygous for the BBS1 M390R mutation. METHODS: Three patients [PT1, F, 27 years old (yo) at last examination, 14-year follow-up (F/U) PT2, F, 15-yo PT3, M, 15-yo, both 1-year F/U] underwent eye exams, Goldmann visual fields (GVFs), dark- (DA) and light-adapted (LA) electroretinograms (ERGs), spectral domain optical coherence tomography (SD-OCT) and fundus autofluorescence (FAF). Vision and systemic history were also collected. RESULTS: All patients had night blindness, hyperopic astigmatism, ptosis or mild blepharospasm, foot polydactyly, 5th finger clinodactyly, history of headaches, and variable, diet-responsive obesity. Two had asthma, PT1 was developmentally delayed, PT2 had Asperger-like symptoms, and PT3 had normal cognition. At age 14, acuity was 20/100 in PT1, who had nystagmus since age 2, 20/40 in PT2 and 20/30 in PT3. By 27yo PT1 progressed to 20/320, by 15 yo PT2 was 20/60 and PT3 remained stable. PT1 had well preserved peripheral GVFs, with minimal progression over 10 years of F/U. PT2 and PT3 presented with ring scotomas and I4e<5°. All patients had severe generalized visual sensitivity depression. ERGs were consistently recordable (also rod ERG in PT3 after 60 min DA), but progressed to non-recordable in PT1. Mixed DA ERGs exhibited electronegativity. In PT3, this was partly due to a bleaching effect during bright-flash DA averaging, partly to ON≫OFF LA response compromise. PT2 and 3 had, on SD-OCTs, generalized macular thinning, normal retinal lamination, and widespread photoreceptor outer/inner segment attenuation except foveally, and multiple rings of abnormal FAF configuring a complex bull's eye-pattern. PT1 had macular atrophy. All patients also had peripapillary nerve fiber layer thickening. CONCLUSIONS: The observed phenotype matches very closely that reported in patients by Azari et al. (IOVS 2006) and in the Bbs1-M390R knock-in mouse model, and expands it to the characterization of important ERG response characteristics that provide insight in the pathogenesis of retinopathy in these patients. Our findings confirm the consistent pathogenicity of the BBS1 M390R mutation.


Subject(s)
Bardet-Biedl Syndrome/genetics , Homozygote , Microtubule-Associated Proteins/genetics , Mutation/genetics , Retinal Diseases/physiopathology , Adolescent , Adult , Bardet-Biedl Syndrome/physiopathology , Electroretinography , Female , Fluorescein Angiography , Humans , Male , Phenotype , Retinal Diseases/genetics , Tomography, Optical Coherence , Visual Fields/physiology
14.
Proc Natl Acad Sci U S A ; 108(51): 20678-83, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22139371

ABSTRACT

Bardet-Biedl syndrome (BBS) is a heterogeneous disorder characterized by obesity, retinopathy, polydactyly, and congenital anomalies. The incidence of hypertension and diabetes are also increased in BBS patients. Mutation of 16 genes independently causes BBS, and seven BBS proteins form the BBSome that promotes ciliary membrane elongation. BBS3 (ARL6), an ADP ribosylation factor-like small GTPase, is not part of the BBSome complex. The in vivo function of BBS3 is largely unknown. Here we developed a Bbs3 knockout model and demonstrate that Bbs3(-/-) mice develop BBS-associated phenotypes, including retinal degeneration, male infertility, and increased body fat. Interestingly, Bbs3(-/-) mice develop some unique phenotypes not seen in other BBS knockout models: no overt obesity, severe hydrocephalus, and elevated blood pressure (shared by some but not all BBS gene knockout mice). We found that endogenous BBS3 and the BBSome physically interact and depend on each other for their ciliary localization. This finding explains the phenotypic similarity between Bbs3(-/-) mice and BBSome subunit knockout mice. Loss of Bbs3 does not affect BBSome formation but disrupts normal localization of melanin concentrating hormone receptor 1 to ciliary membranes and affects retrograde transport of Smoothened inside cilia. We also show that the endogenous BBSome and BBS3 associate with membranes and the membrane association of the BBSome and BBS3 are not interdependent. Differences between BBS mouse models suggest nonoverlapping functions to individual BBS protein.


Subject(s)
ADP-Ribosylation Factors/genetics , Bardet-Biedl Syndrome/genetics , Mutation , ADP-Ribosylation Factors/physiology , Animals , Brain/metabolism , Exons , Flagella/metabolism , Homozygote , Humans , Male , Mice , Mice, Knockout , Microtubules/metabolism , Obesity/metabolism , Phenotype , Protein Transport , Spermatozoa/physiology
15.
J Clin Invest ; 121(9): 3542-53, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21821918

ABSTRACT

Mutations in myocilin (MYOC) are the most common genetic cause of primary open angle glaucoma (POAG), but the mechanisms underlying MYOC-associated glaucoma are not fully understood. Here, we report the development of a transgenic mouse model of POAG caused by the Y437H MYOC mutation; the mice are referred to herein as Tg-MYOC(Y437H) mice. Analysis of adult Tg-MYOC(Y437H) mice, which we showed express human MYOC containing the Y437H mutation within relevant eye tissues, revealed that they display glaucoma phenotypes (i.e., elevated intraocular pressure [IOP], retinal ganglion cell death, and axonal degeneration) closely resembling those seen in patients with POAG caused by the Y437H MYOC mutation. Mutant myocilin was not secreted into the aqueous humor but accumulated in the ER of the trabecular meshwork (TM), thereby inducing ER stress in the TM of Tg-MYOC(Y437H) mice. Furthermore, chronic and persistent ER stress was found to be associated with TM cell death and elevation of IOP in Tg-MYOC(Y437H) mice. Reduction of ER stress with a chemical chaperone, phenylbutyric acid (PBA), prevented glaucoma phenotypes in Tg-MYOC(Y437H) mice by promoting the secretion of mutant myocilin in the aqueous humor and by decreasing intracellular accumulation of myocilin in the ER, thus preventing TM cell death. These results demonstrate that ER stress is linked to the pathogenesis of POAG and may be a target for treatment in human patients.


Subject(s)
Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Glaucoma, Open-Angle/drug therapy , Glaucoma, Open-Angle/physiopathology , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , Stress, Physiological , Animals , Apoptosis/physiology , Cells, Cultured , Cytoskeletal Proteins/genetics , Endoplasmic Reticulum/pathology , Eye Proteins/genetics , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/pathology , Glycoproteins/genetics , Humans , Intraocular Pressure , Mice , Mice, Transgenic , Mutation , Phenotype , Trabecular Meshwork/cytology , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology , Transgenes , Unfolded Protein Response
16.
Am J Physiol Renal Physiol ; 300(2): F574-80, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21106857

ABSTRACT

Bardet-Biedl syndrome (BBS) is a rare hereditary autosomal recessive disease associated with several features including obesity, hypertension, and renal abnormalities. The underlying mechanisms of renal defects associated with BBS remain poorly defined. We examined the histological, molecular, and functional renal changes in BBS mouse models that have features of the human disorder. Interestingly, obese hypertensive Bbs4(-/-) mice exhibited inflammatory infiltration and renal cysts, whereas the obese normotensive Bbs2(-/-) mice had only minor inflammatory infiltration. Accordingly, the expression level of inducible nitric oxide synthase was elevated in the kidney of both BBS mice with a more marked increase in Bbs4(-/-) mice. In contrast, endothelial nitric oxide synthase expression was decreased in Bbs4(-/-), but not Bbs2(-/-), mice. Similarly, the expression levels of transient receptor potential vanilloid 1 and 4 channels as well as ß- and γ-subunits of epithelial Na channel were significantly reduced only in the kidney of Bbs4(-/-) mice. Metabolic studies revealed changes in urine output and urinary concentrations of creatinine, blood urea nitrogen, sodium, and potassium with a more pronounced effect in Bbs4(-/-) mice. Finally, we found that calorie restriction which prevented obesity in BBS mice reversed the morphological and molecular changes found in Bbs2(-/-) and Bbs4(-/-) mice, indicating the kidney abnormalities associated with BBS are obesity related. These findings extend our understanding of the function of BBS proteins and emphasize the importance of these proteins in renal physiology.


Subject(s)
Bardet-Biedl Syndrome/genetics , Kidney/abnormalities , Microtubule-Associated Proteins/genetics , Proteins/genetics , Animals , Blood Urea Nitrogen , Caloric Restriction , Creatinine/urine , Epithelial Sodium Channels/analysis , Female , Hypertension/genetics , Male , Mice , Nitric Oxide Synthase Type III/analysis , Obesity/genetics , Oliguria/physiopathology , Potassium/urine , Sodium/urine , TRPV Cation Channels/analysis
17.
Genet Med ; 12(10): 623-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20949666

ABSTRACT

PURPOSE: Bardet-Biedl syndrome is a pleiotropic multiple anomaly syndrome inherited in an autosomal recessive pattern. It is now known that this disorder has locus heterogeneity, with causative mutations identified in as many as 14 genes. The aim of this study was to derive locus-specific recurrence risk estimates for family members of a proband affected with Bardet-Biedl syndrome. METHODS: Mutation data from 187 probands affected with Bardet-Biedl syndrome were used. The authors counted the relative proportion of families with mutations at each of 10 loci and estimated locus-specific carrier rates for mutations using Hardy-Weinberg principles and an aggregate population frequency of 1/100,000 for the phenotype. Locus-specific recurrence risks were calculated for relatives of an affected proband. RESULTS: Locus-specific carrier frequencies range from 1/250 to 1/2200, and the risks for an offspring of the sibling of an affected individual range from 1/1,500 to 1/13,000. The estimate of this risk derived under a locus homogeneity model is 1/960. CONCLUSION: Variation of recurrence risks of this magnitude may have implications for genetic counseling of families with affected individuals, in particular about prenatal testing and other reproductive options. Similar analyses to determine locus-specific carrier frequencies for other phenotypes with significant locus heterogeneity may yield similarly relevant results.


Subject(s)
Bardet-Biedl Syndrome/diagnosis , Bardet-Biedl Syndrome/genetics , DNA Mutational Analysis , Genetic Carrier Screening , Genetic Heterogeneity , Abnormalities, Multiple/genetics , Alleles , Gene Frequency , Genetic Loci , Genetic Testing , Heterozygote , Humans , Models, Genetic , Mutation , Obesity , Pedigree , Prenatal Diagnosis , Retinitis Pigmentosa , Risk Assessment
18.
Am J Hum Genet ; 86(5): 686-95, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20398886

ABSTRACT

Retinitis pigmentosa is a genetically heterogeneous group of inherited ocular disorders characterized by progressive photoreceptor cell loss, night blindness, constriction of the visual field, and progressive visual disability. Homozygosity mapping and gene expression studies identified a 2 exon gene, C2ORF71. The encoded protein has no homologs and is highly expressed in the eye, where it is specifically expressed in photoreceptor cells. Two mutations were found in C2ORF71 in human RP patients: A nonsense mutation (p.W253X) in the first exon is likely to be a null allele; the second, a missense mutation (p.I201F) within a highly conserved region of the protein, leads to proteosomal degradation. Bioinformatic and functional studies identified and validated sites of lipid modification within the first three amino acids of the C2ORF71 protein. Using morpholino oligonucleotides to knockdown c2orf71 expression in zebrafish results in visual defects, confirming that C2ORF71 plays an important role in the development of normal vision. Finally, localization of C2ORF71 to primary cilia in cultured cells suggests that the protein is likely to localize to the connecting cilium or outer segment of photoreceptor cells.


Subject(s)
Eye/metabolism , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Proteins/genetics , Retinitis Pigmentosa/genetics , Blindness/genetics , Cilia/genetics , Cilia/metabolism , Exons , Eye Proteins/genetics , Homozygote , Humans , Mutation, Missense , Retinitis Pigmentosa/metabolism
19.
PLoS Genet ; 6(3): e1000884, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333246

ABSTRACT

Bardet-Biedl Syndrome (BBS) is a heterogeneous syndromic form of retinal degeneration. We have identified a novel transcript of a known BBS gene, BBS3 (ARL6), which includes an additional exon. This transcript, BBS3L, is evolutionally conserved and is expressed predominantly in the eye, suggesting a specialized role in vision. Using antisense oligonucleotide knockdown in zebrafish, we previously demonstrated that bbs3 knockdown results in the cardinal features of BBS in zebrafish, including defects to the ciliated Kupffer's Vesicle and delayed retrograde melanosome transport. Unlike bbs3, knockdown of bbs3L does not result in Kupffer's Vesicle or melanosome transport defects, rather its knockdown leads to impaired visual function and mislocalization of the photopigment green cone opsin. Moreover, BBS3L RNA, but not BBS3 RNA, is sufficient to rescue both the vision defect as well as green opsin localization in the zebrafish retina. In order to demonstrate a role for Bbs3L function in the mammalian eye, we generated a Bbs3L-null mouse that presents with disruption of the normal photoreceptor architecture. Bbs3L-null mice lack key features of previously published Bbs-null mice, including obesity. These data demonstrate that the BBS3L transcript is required for proper retinal function and organization.


Subject(s)
ADP-Ribosylation Factors/metabolism , Vision, Ocular , Zebrafish Proteins/metabolism , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/deficiency , ADP-Ribosylation Factors/genetics , Amino Acid Sequence , Animals , Bardet-Biedl Syndrome/complications , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/pathology , Bardet-Biedl Syndrome/physiopathology , Eye Abnormalities/complications , Eye Abnormalities/pathology , Eye Abnormalities/physiopathology , Ganglia/drug effects , Ganglia/metabolism , Ganglia/pathology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Mice , Mice, Mutant Strains , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , Organ Specificity/drug effects , Phenotype , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reflex, Startle/drug effects , Rod Opsins/metabolism , Vision, Ocular/drug effects , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
20.
Hum Mutat ; 31(4): 429-36, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20120035

ABSTRACT

Bardet-Biedl syndrome (BBS) is an autosomal recessive disease characterized by retinal dystrophy, polydactyly, obesity, learning disabilities, renal involvement, and male hypogenitalism. BBS is genetically heterogeneous with mutations of 14 genes, accounting for approximately 70% of cases. Triallelic inheritance has been suggested in about 5% of cases. Forty-nine unrelated BBS patients were screened for mutations by DHPLC analysis in BBS1, BBS2, BBS4, BBS6/MKKS, BBS10, and BBS12. The selected genes either account for more than 5% of the mutational load or are commonly reported in triallelic inheritance. Eight patients with only one or no BBS mutation were further investigated by single nucleotide polymorphism (SNP) analysis. In total, mutations were detected in 44 patients. Twenty percent had two mutations in BBS1, 18% in BBS2, 4% in BBS9, 43% in BBS10, and 2% in BBS12. Five patients were heterozygous for a sequence variation in BBS6/MKKS. We found eight patients with three sequence variations in two genes, which could be explained by triallelic inheritance, by the prevalence of heterozygous carriers or the third sequence variations representing rare polymorphisms. All changes found in a second BBS gene were amino acid substitutions. Genotype-phenotype correlations suggest a milder phenotype for BBS1 compared to BBS2 and BBS10, which we ascribe to the hypomorphic p.Met390Arg-mutation.


Subject(s)
Bardet-Biedl Syndrome/genetics , Mutation/genetics , Alleles , Amino Acid Sequence , Base Sequence , Conserved Sequence , DNA Mutational Analysis , Denmark , Evolution, Molecular , Female , Genetic Association Studies , Genotype , Group II Chaperonins/chemistry , Group II Chaperonins/genetics , Humans , Inheritance Patterns/genetics , Male , Molecular Sequence Data , Polymorphism, Single Nucleotide/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...