Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38576540

ABSTRACT

Aging is the most significant risk factor for age-related diseases in general, which is true for age-related diseases in the eye including age-related macular degeneration (AMD). Therefore, in order to identify potential therapeutic targets for these diseases, it is crucial to understand the normal aging process and how its mis-regulation could cause age-related diseases at the molecular level. Recently, abnormal lipid metabolism has emerged as one major aspect of age-related symptoms in the retina. Animal models provide excellent means to identify and study factors that regulate lipid metabolism in relation to age-related symptoms. Central to this review is the role of transmembrane protein 135 (TMEM135) in the retina. TMEM135 was identified through the characterization of a mutant mouse strain exhibiting accelerated retinal aging and positional cloning of the responsible mutation within the gene, indicating the crucial role of TMEM135 in regulating the normal aging process in the retina. Over the past decade, the molecular functions of TMEM135 have been explored in various models and tissues, providing insights into the regulation of metabolism, particularly lipid metabolism, through its action in multiple organelles. Studies indicated that TMEM135 is a significant regulator of peroxisomes, mitochondria, and their interaction. Here, we provide an overview of the molecular functions of TMEM135 which is crucial for regulating mitochondria, peroxisomes, and lipids. The review also discusses the age-dependent phenotypes in mice with TMEM135 perturbations, emphasizing the importance of a balanced TMEM135 function for the health of the retina and other tissues including the heart, liver, and adipose tissue. Finally, we explore the potential roles of TMEM135 in human age-related retinal diseases, connecting its functions to the pathobiology of AMD.

2.
J Vis Exp ; (193)2023 03 10.
Article in English | MEDLINE | ID: mdl-36971449

ABSTRACT

Age-related macular degeneration (AMD) is a debilitating retinal disorder in aging populations. It is widely believed that dysfunction of the retinal pigmented epithelium (RPE) is a key pathobiological event in AMD. To understand the mechanisms that lead to RPE dysfunction, mouse models can be utilized by researchers. It has been established by previous studies that mice can develop RPE pathologies, some of which are observed in the eyes of individuals diagnosed with AMD. Here, we describe a phenotyping protocol to assess RPE pathologies in mice. This protocol includes the preparation and evaluation of retinal cross-sections using light microscopy and transmission electron microscopy, as well as that of RPE flat mounts by confocal microscopy. We detail the common types of murine RPE pathologies observed by these techniques and ways to quantify them through unbiased methods for statistical testing. As proof of concept, we use this RPE phenotyping protocol to quantify the RPE pathologies observed in mice overexpressing transmembrane protein 135 (Tmem135) and aged wild-type C57BL/6J mice. The main goal of this protocol is to present standard RPE phenotyping methods with unbiased quantitative assessments for scientists using mouse models of AMD.


Subject(s)
Macular Degeneration , Mice , Animals , Mice, Inbred C57BL , Macular Degeneration/pathology , Retinal Pigment Epithelium/metabolism , Retina/metabolism , Disease Models, Animal , Epithelium/metabolism
3.
Commun Biol ; 6(1): 8, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36599953

ABSTRACT

Transmembrane protein 135 (TMEM135) is thought to participate in the cellular response to increased intracellular lipids yet no defined molecular function for TMEM135 in lipid metabolism has been identified. In this study, we performed a lipid analysis of tissues from Tmem135 mutant mice and found striking reductions of docosahexaenoic acid (DHA) across all Tmem135 mutant tissues, indicating a role of TMEM135 in the production of DHA. Since all enzymes required for DHA synthesis remain intact in Tmem135 mutant mice, we hypothesized that TMEM135 is involved in the export of DHA from peroxisomes. The Tmem135 mutation likely leads to the retention of DHA in peroxisomes, causing DHA to be degraded within peroxisomes by their beta-oxidation machinery. This may lead to generation or alteration of ligands required for the activation of peroxisome proliferator-activated receptor a (PPARa) signaling, which in turn could result in increased peroxisomal number and beta-oxidation enzymes observed in Tmem135 mutant mice. We confirmed this effect of PPARa signaling by detecting decreased peroxisomes and their proteins upon genetic ablation of Ppara in Tmem135 mutant mice. Using Tmem135 mutant mice, we also validated the protective effect of increased peroxisomes and peroxisomal beta-oxidation on the metabolic disease phenotypes of leptin mutant mice which has been observed in previous studies. Thus, we conclude that TMEM135 has a role in lipid homeostasis through its function in peroxisomes.


Subject(s)
Docosahexaenoic Acids , Lipid Metabolism , Membrane Proteins , Peroxisomes , Animals , Mice , Docosahexaenoic Acids/metabolism , Homeostasis , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisomes/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism
4.
Sci Rep ; 12(1): 756, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35031662

ABSTRACT

Aging is a significant factor in the development of age-related diseases but how aging disrupts cellular homeostasis to cause age-related retinal disease is unknown. Here, we further our studies on transmembrane protein 135 (Tmem135), a gene involved in retinal aging, by examining the transcriptomic profiles of wild-type, heterozygous and homozygous Tmem135 mutant posterior eyecup samples through RNA sequencing (RNA-Seq). We found significant gene expression changes in both heterozygous and homozygous Tmem135 mutant mouse eyecups that correlate with visual function deficits. Further analysis revealed that expression of many genes involved in lipid metabolism are changed due to the Tmem135 mutation. Consistent with these changes, we found increased lipid accumulation in mutant Tmem135 eyecup samples. Since mutant Tmem135 mice have similar ocular pathologies as human age-related macular degeneration (AMD) eyes, we compared our homozygous Tmem135 mutant eyecup RNA-Seq dataset with transcriptomic datasets of human AMD donor eyes. We found similar changes in genes involved in lipid metabolism between the homozygous Tmem135 mutant eyecups and AMD donor eyes. Our study suggests that the Tmem135 mutation affects lipid metabolism as similarly observed in human AMD eyes, thus Tmem135 mutant mice can serve as a good model for the role of dysregulated lipid metabolism in AMD.


Subject(s)
Eye/metabolism , Lipid Metabolism/genetics , Macular Degeneration/etiology , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Mutation , Animals , Disease Models, Animal , Humans , Macular Degeneration/genetics , Mice, Mutant Strains
5.
J Ocul Pharmacol Ther ; 38(1): 3-32, 2022.
Article in English | MEDLINE | ID: mdl-34788573

ABSTRACT

Age-related macular degeneration (AMD) is a major leading cause of irreversible visual impairment in the world with limited therapeutic interventions. Histological, biochemical, genetic, and epidemiological studies strongly implicate dysregulated lipid metabolism in the retinal pigmented epithelium (RPE) in AMD pathobiology. However, effective therapies targeting lipid metabolism still need to be identified and developed for this blinding disease. To test lipid metabolism-targeting therapies, preclinical AMD mouse models are needed to establish therapeutic efficacy and the role of lipid metabolism in the development of AMD-like pathology. In this review, we provide a comprehensive overview of current AMD mouse models available to researchers that could be used to provide preclinical evidence supporting therapies targeting lipid metabolism for AMD. Based on previous studies of AMD mouse models, we discuss strategies to modulate lipid metabolism as well as examples of studies evaluating lipid-targeting therapeutics to restore lipid processing in the RPE. The use of AMD mouse models may lead to worthy lipid-targeting candidate therapies for clinical trials to prevent the blindness caused by AMD.


Subject(s)
Disease Models, Animal , Lipid Metabolism/physiology , Macular Degeneration/physiopathology , Animals , Macular Degeneration/genetics , Mice , Risk Factors
6.
Invest Ophthalmol Vis Sci ; 61(12): 16, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33064130

ABSTRACT

Purpose: Aging is a critical risk factor for the development of retinal diseases, but how aging perturbs ocular homeostasis and contributes to disease is unknown. We identified transmembrane protein 135 (Tmem135) as a gene important for regulating retinal aging and mitochondrial dynamics in mice. Overexpression of Tmem135 causes mitochondrial fragmentation and pathologies in the hearts of mice. In this study, we examine the eyes of mice overexpressing wild-type Tmem135 (Tmem135 TG) and compare their phenotype to Tmem135 mutant mice. Methods: Eyes were collected for histology, immunohistochemistry, electron microscopy, quantitative PCR, and Western blot analysis. Before tissue collection, electroretinography (ERG) was performed to assess visual function. Mouse retinal pigmented epithelium (RPE) cultures were established to visualize mitochondria. Results: Pathologies were observed only in the RPE of Tmem135 TG mice, including degeneration, migratory cells, vacuolization, dysmorphogenesis, cell enlargement, and basal laminar deposit formation despite similar augmented levels of Tmem135 in the eyecup (RPE/choroid/sclera) and neural retina. We observed reduced mitochondria number and size in the Tmem135 TG RPE. ERG amplitudes were decreased in 365-day-old mice overexpressing Tmem135 that correlated with reduced expression of RPE cell markers. In Tmem135 mutant mice, RPE cells are thicker, smaller, and denser than their littermate controls without any signs of degeneration. Conclusions: Overexpression and mutation of Tmem135 cause contrasting RPE abnormalities in mice that correlate with changes in mitochondrial shape and size (overfragmented in TG vs. overfused in mutant). We conclude proper regulation of mitochondrial homeostasis by TMEM135 is critical for RPE health.


Subject(s)
Gene Expression Regulation/physiology , Membrane Proteins/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mutation/genetics , Retinal Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Animals , Blotting, Western , Cell Count , Cells, Cultured , Disease Models, Animal , Electroretinography , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology , Phenotype , Real-Time Polymerase Chain Reaction , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/pathology
7.
J Biol Chem ; 295(39): 13601-13616, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32737203

ABSTRACT

Strong evidence suggests that dysregulated lipid metabolism involving dysfunction of the retinal pigmented epithelium (RPE) underlies the pathogenesis of age-related macular degeneration (AMD), the leading cause of irreversible blindness in the elderly. A hallmark of AMD is the overproduction of lipid- and protein-rich extracellular deposits that accumulate in the extracellular matrix (Bruch's membrane (BrM)) adjacent to the RPE. We analyzed apolipoprotein A-1 (ApoA-1)-containing lipoproteins isolated from BrM of elderly human donor eyes and found a unique proteome, distinct from high-density lipoprotein (HDL) isolated from donor plasma of the same individuals. The most striking difference is higher concentrations of ApoB and ApoE, which bind to glycosaminoglycans. We hypothesize that this interaction promotes lipoprotein deposition onto BrM glycosaminoglycans, initiating downstream effects that contribute to RPE dysfunction/death. We tested this hypothesis using two potential therapeutic strategies to alter the lipoprotein/protein profile of these extracellular deposits. First, we used short heparan sulfate oligosaccharides to remove lipoproteins already deposited in both the extracellular matrix of RPE cells and aged donor BrM tissue. Second, an ApoA-1 mimetic, 5A peptide, was demonstrated to modulate the composition and concentration of apolipoproteins secreted from primary porcine RPE cells. Significantly, in a mouse model of AMD, this 5A peptide altered the proteomic profile of circulating HDL and ameliorated some of the potentially harmful changes to the protein composition resulting from the high-fat, high-cholesterol diet in this model. Together, these results suggest that targeting HDL interactions with BrM represents a new strategy to slow AMD progression in humans.


Subject(s)
Lipoproteins, HDL/metabolism , Macular Degeneration/metabolism , Animals , Apolipoprotein A-I/analysis , Apolipoprotein A-I/metabolism , Bruch Membrane/metabolism , Cells, Cultured , Humans , Lipoproteins, HDL/blood , Lipoproteins, HDL/isolation & purification , Mice , Retinal Pigment Epithelium/metabolism , Swine
8.
Genetics ; 214(1): 121-134, 2020 01.
Article in English | MEDLINE | ID: mdl-31754016

ABSTRACT

One major aspect of the aging process is the onset of chronic, low-grade inflammation that is highly associated with age-related diseases. The molecular mechanisms that regulate these processes have not been fully elucidated. We have identified a spontaneous mutant mouse line, small with kinky tail (skt), that exhibits accelerated aging and age-related disease phenotypes including increased inflammation in the brain and retina, enhanced age-dependent retinal abnormalities including photoreceptor cell degeneration, neurodegeneration in the hippocampus, and reduced lifespan. By positional cloning, we identified a deletion in chondroitin sulfate synthase 1 (Chsy1) that is responsible for these phenotypes in skt mice. CHSY1 is a member of the chondroitin N-acetylgalactosaminyltransferase family that plays critical roles in the biosynthesis of chondroitin sulfate, a glycosaminoglycan (GAG) that is attached to the core protein to form the chondroitin sulfate proteoglycan (CSPG). Consistent with this function, the Chsy1 mutation dramatically decreases chondroitin sulfate GAGs in the retina and hippocampus. In addition, macrophage and neutrophil populations appear significantly altered in the bone marrow and spleen of skt mice, suggesting an important role for CHSY1 in the functioning of these immune cell types. Thus, our study reveals a previously unidentified impact of CHSY1 in the retina and hippocampus. Specifically, chondroitin sulfate (CS) modification of proteins by CHSY1 appears critical for proper regulation of immune cells of the myeloid lineage and for maintaining the integrity of neuronal tissues, since a defect in this gene results in increased inflammation and abnormal phenotypes associated with age-related diseases.


Subject(s)
Chondroitin Sulfates/metabolism , Glucuronosyltransferase/metabolism , Inflammation/metabolism , Multifunctional Enzymes/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Neurodegenerative Diseases/metabolism , Protein Processing, Post-Translational , Proteins/genetics , Retinal Degeneration/metabolism , Age Factors , Animals , Apoptosis/physiology , Female , Glucuronosyltransferase/genetics , Inflammation/genetics , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multifunctional Enzymes/genetics , Mutation , N-Acetylgalactosaminyltransferases/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Proteins/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/pathology
9.
Proc Natl Acad Sci U S A ; 116(9): 3703-3711, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808757

ABSTRACT

One of the strongest susceptibility genes for age-related macular degeneration (AMD) is complement factor H (CFH); however, its impact on AMD pathobiology remains unresolved. Here, the effect of the principal AMD-risk-associated CFH variant (Y402H) on the development and progression of age-dependent AMD-like pathologies was determined in vivo. Transgenic mice expressing equal amounts of the full-length normal human CFH Y402 (CFH-Y/0) or the AMD-risk associated CFH H402 (CFH-H/H) variant on a Cfh-/- background were aged to 90 weeks and switched from normal diet (ND) to a high fat, cholesterol-enriched (HFC) diet for 8 weeks. The resulting phenotype was compared with age-matched controls maintained on ND. Remarkably, an AMD-like phenotype consisting of vision loss, increased retinal pigmented epithelium (RPE) stress, and increased basal laminar deposits was detected only in aged CFH-H/H mice following the HFC diet. These changes were not observed in aged CFH-Y/0 mice or in younger (36- to 40-week-old) CFH mice of both genotypes fed either diet. Biochemical analyses of aged CFH mice after HFC diet revealed genotype-dependent changes in plasma and eyecup lipoproteins, but not complement activation, which correlated with the AMD-like phenotype in old CFH-H/H mice. Specifically, apolipoproteins B48 and A1 are elevated in the RPE/choroid of the aged CFH-H/H mice compared with age-matched control CFH-Y/0 fed a HFC diet. Hence, we demonstrate a functional consequence of the Y402H polymorphism in vivo, which promotes AMD-like pathology development and affects lipoprotein levels in aged mice. These findings support targeting lipoproteins as a viable therapeutic strategy for treating AMD.


Subject(s)
Complement Activation/genetics , Complement Factor H/genetics , Lipoproteins/genetics , Macular Degeneration/genetics , Animals , Diet, High-Fat/adverse effects , Female , Genotype , Humans , Lipoproteins/metabolism , Macular Degeneration/pathology , Male , Mice , Mice, Transgenic/genetics , Polymorphism, Single Nucleotide/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
10.
Invest Ophthalmol Vis Sci ; 59(2): 662-673, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29392311

ABSTRACT

Purpose: A large body of evidence supports a central role for complement activation in the pathobiology of age-related macular degeneration (AMD), including plasma complement component 5a (C5a). Interestingly, C5a is a chemotactic agent for monocytes, a cell type also shown to contribute to AMD. However, the role monocytes play in the pathogenesis of "dry" AMD and the pharmacologic potential of targeting C5a to regulate these cells are unclear. We addressed these questions via C5a blockade in a unique model of early/intermediate dry AMD and large panel flow cytometry to immunophenotype monocytic involvement. Methods: Heterozygous complement factor H (Cfh+/-) mice aged to 90 weeks were fed a high-fat, cholesterol-enriched diet (Cfh+/-∼HFC) for 8 weeks and were given weekly intraperitoneal injections of 30 mg/kg anti-C5a (4C9, Pfizer). Flow cytometry, retinal pigmented epithelium (RPE) flat mounts, and electroretinograms were used to characterize anti-C5a treatment. Results: Aged Cfh+/- mice developed RPE damage, sub-RPE basal laminar deposits, and attenuation of visual function and immune cell recruitment to the choroid that was accompanied by expression of inflammatory and extracellular matrix remodeling genes following 8 weeks of HFC diet. Concomitant systemic administration of an anti-C5a antibody successfully inhibited local recruitment of mononuclear phagocytes to the choroid-RPE interface but did not ameliorate these AMD-like pathologies in this mouse model. Conclusions: These results show that immunotherapy targeting C5a is not sufficient to block the development of the AMD-like pathologies observed in Cfh+/-∼HFC mice and suggest that other complement components or molecules/mechanisms may be driving "early" and "intermediate" AMD pathologies.


Subject(s)
Antibodies, Blocking/therapeutic use , Choroidal Neovascularization/therapy , Complement C5a/antagonists & inhibitors , Disease Models, Animal , Geographic Atrophy/therapy , Immunotherapy , Animals , Cholesterol, Dietary/administration & dosage , Choroidal Neovascularization/immunology , Choroidal Neovascularization/pathology , Complement Activation , Complement C5a/immunology , Electroretinography , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Retinal Pigment Epithelium/pathology
11.
Am J Pathol ; 185(1): 29-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25447048

ABSTRACT

Complement factor H (CFH) is an important regulatory protein in the alternative pathway of the complement system, and CFH polymorphisms increase the genetic risk of age-related macular degeneration dramatically. These same human CFH variants have also been associated with dense deposit disease. To mechanistically study the function of CFH in the pathogenesis of these diseases, we created transgenic mouse lines using human CFH bacterial artificial chromosomes expressing full-length human CFH variants and crossed these to Cfh knockout (Cfh(-/-)) mice. Human CFH protein inhibited cleavage of mouse complement component 3 and factor B in plasma and in retinal pigment epithelium/choroid/sclera, establishing that human CFH regulates activation of the mouse alternative pathway. One of the mouse lines, which express relatively higher levels of CFH, demonstrated functional and structural protection of the retina owing to the Cfh deletion. Impaired visual function, detected as a deficit in the scotopic electroretinographic response, was improved in this transgenic mouse line compared with Cfh(-/-) mice, and transgenics had a thicker outer nuclear layer and less sub-retinal pigment epithelium deposit accumulation. In addition, expression of human CFH also completely protected the mice from developing kidney abnormalities associated with loss of CFH. These humanized CFH mice present a valuable model for study of the molecular mechanisms of age-related macular degeneration and dense deposit disease and for testing therapeutic targets.


Subject(s)
Kidney Diseases/genetics , Macular Degeneration/genetics , Retinal Diseases/genetics , Animals , Choroid/pathology , Complement C3/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Crosses, Genetic , Electroretinography , Humans , Kidney Diseases/pathology , Macular Degeneration/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Retina/metabolism , Retinal Diseases/pathology , Retinal Pigment Epithelium/pathology , Sclera/pathology , Sheep
12.
Blood ; 122(14): 2487-90, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23943650

ABSTRACT

Classical 5q- syndrome is an acquired macrocytic anemia of the elderly. Similar to Diamond Blackfan anemia (DBA), an inherited red cell aplasia, the bone marrow is characterized by a paucity of erythroid precursors. RPS14 deletions in combination with other deletions in the region have been implicated as causative of the 5q- syndrome phenotype. We asked whether smaller, less easily detectable deletions could account for a syndrome with a modified phenotype. We employed single-nucleotide polymorphism array genotyping to identify small deletions in patients diagnosed with DBA and other anemias lacking molecular diagnoses. Diminutive mosaic deletions involving RPS14 were identified in a 5-year-old patient with nonclassical DBA and in a 17-year-old patient with myelodysplastic syndrome. Patients with nonclassical DBA and other hypoproliferative anemias may have somatically acquired 5q deletions with RPS14 haploinsufficiency not identified by fluorescence in situ hybridization or cytogenetic testing, thus refining the spectrum of disorders with 5q- deletions.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Macrocytic/genetics , Cytogenetic Analysis/methods , Ribosomal Proteins/genetics , Adolescent , Anemia, Diamond-Blackfan/diagnosis , Anemia, Macrocytic/diagnosis , Anemia, Macrocytic/drug therapy , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Female , Genotype , Humans , Immunologic Factors/therapeutic use , Lenalidomide , Phenotype , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use
13.
Hum Genet ; 132(11): 1265-74, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23812780

ABSTRACT

Diamond-Blackfan anemia (DBA) is an inherited red blood cell aplasia that usually presents during the first year of life. The main features of the disease are normochromic and macrocytic anemia, reticulocytopenia, and nearly absent erythroid progenitors in the bone marrow. The patients also present with growth retardation and craniofacial, upper limb, heart and urinary system congenital malformations in ~30-50 % of cases. The disease has been associated with point mutations and large deletions in ten ribosomal protein (RP) genes RPS19, RPS24, RPS17, RPL35A, RPL5, RPL11, RPS7, RPS10, RPS26, and RPL26 and GATA1 in about 60-65 % of patients. Here, we report a novel large deletion in RPL15, a gene not previously implicated to be causative in DBA. Like RPL26, RPL15 presents the distinctive feature of being required both for 60S subunit formation and for efficient cleavage of the internal transcribed spacer 1. In addition, we detected five deletions in RP genes in which mutations have been previously shown to cause DBA: one each in RPS19, RPS24, and RPS26, and two in RPS17. Pre-ribosomal RNA processing was affected in cells established from the patients bearing these deletions, suggesting a possible molecular basis for their pathological effect. These data identify RPL15 as a new gene involved in DBA and further support the presence of large deletions in RP genes in DBA patients.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Gene Deletion , Ribosomal Proteins/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations , Gene Knockdown Techniques , HeLa Cells , Humans , Mutation , RNA, Ribosomal/analysis , RNA, Ribosomal/genetics , RNA, Small Interfering , Ribosomal Proteins/metabolism
14.
Hum Mutat ; 33(7): 1037-44, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22431104

ABSTRACT

Diamond-Blackfan anemia (DBA) is an inherited form of pure red cell aplasia that usually presents in infancy or early childhood and is associated with congenital malformations in ∼30-50% of patients. DBA has been associated with mutations in nine ribosomal protein (RP) genes in about 53% of patients. We completed a large-scale screen of 79 RP genes by sequencing 16 RP genes (RPL3, RPL7, RPL8, RPL10, RPL14, RPL17, RPL19, RPL23A, RPL26, RPL27, RPL35, RPL36A, RPL39, RPS4X, RPS4Y1, and RPS21) in 96 DBA probands. We identified a de novo two-nucleotide deletion in RPL26 in one proband associated with multiple severe physical abnormalities. This mutation gives rise to a remarkable ribosome biogenesis defect that affects maturation of both the small and the large subunits. We also found a deletion in RPL19 and missense mutations in RPL3 and RPL23A, which may be variants of unknown significance. Together with RPL5, RPL11, and RPS7, RPL26 is the fourth RP regulating p53 activity that is linked to DBA.


Subject(s)
Abnormalities, Multiple/genetics , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Frameshift Mutation/genetics , RNA, Ribosomal/genetics , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Blotting, Northern , Blotting, Western , HeLa Cells , Humans , RNA, Small Interfering , Ribosomal Protein L3 , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/genetics
15.
J Surg Res ; 171(1): 23-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21571316

ABSTRACT

BACKGROUND: Human medullary thyroid cancer (MTC) is a neuroendocrine (NE) tumor, derived from thyroid C-cells. Besides surgery, there are no curative therapies for MTC. This emphasizes the need for the development of new therapies. In MTC, Notch1 signaling pathway is absent and Notch1 activation in MTC-TT cells has been shown to reduce growth and NE markers in vitro. While the in vitro studies will provide insight into the potential mechanisms by which Notch inhibits growth, only by in vivo model one can recreate the conditions found in patients with MTC and assess effects on metastatic potential and microscopic disease. MATERIALS AND METHODS: Doxycycline inducible TT-NOTCH1 cells were utilized in a murine subcutaneous xenograft model to study tumor development and growth. Doxycycline was used to induce the expression of Notch1 in these tumors. RESULTS: Measurements of tumor volume showed that doxycycline treated mice had slower tumor growth than control mice. Western blot analysis of tumor lysates demonstrated activation of Notch1 protein only in doxycycline treated mice suggesting that active Notch1 slowed tumor growth. Furthermore, this activation led to a significant reduction in the levels of achaete-scute complex-like1 and chromogranin A important NE markers. CONCLUSION: Based on these data, activation of Notch signaling pathway could be a therapeutic strategy to treat patients with MTC.


Subject(s)
Carcinoma, Medullary/pathology , Carcinoma, Neuroendocrine/pathology , Receptor, Notch1/genetics , Thyroid Neoplasms/pathology , Achaete-Scute Complex Genome Region/genetics , Animals , Anti-Bacterial Agents/pharmacology , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Chromogranin A/genetics , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Receptor, Notch1/metabolism , Signal Transduction/physiology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Transplantation, Heterologous
16.
Hum Mutat ; 31(12): 1269-79, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20960466

ABSTRACT

Diamond-Blackfan Anemia (DBA) is characterized by a defect of erythroid progenitors and, clinically, by anemia and malformations. DBA exhibits an autosomal dominant pattern of inheritance with incomplete penetrance. Currently nine genes, all encoding ribosomal proteins (RP), have been found mutated in approximately 50% of patients. Experimental evidence supports the hypothesis that DBA is primarily the result of defective ribosome synthesis. By means of a large collaboration among six centers, we report here a mutation update that includes nine genes and 220 distinct mutations, 56 of which are new. The DBA Mutation Database now includes data from 355 patients. Of those where inheritance has been examined, 125 patients carry a de novo mutation and 72 an inherited mutation. Mutagenesis may be ascribed to slippage in 65.5% of indels, whereas CpG dinucleotides are involved in 23% of transitions. Using bioinformatic tools we show that gene conversion mechanism is not common in RP genes mutagenesis, notwithstanding the abundance of RP pseudogenes. Genotype-phenotype analysis reveals that malformations are more frequently associated with mutations in RPL5 and RPL11 than in the other genes. All currently reported DBA mutations together with their functional and clinical data are included in the DBA Mutation Database.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Databases, Genetic , Mutation/genetics , Ribosomes/genetics , Anemia, Diamond-Blackfan/diagnosis , Base Sequence , Genetic Association Studies , Humans , Molecular Sequence Data , Mutagenesis/genetics , Ribosomal Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...