Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Adv Exp Med Biol ; 1415: 359-363, 2023.
Article in English | MEDLINE | ID: mdl-37440057

ABSTRACT

Age is a major risk factor for age-related macular degeneration (AMD), and age has a role in the disease phenotypes of heritable macular dystrophies. The proteomes of C57Bl6/J mouse choroids at 2 ages were analyzed to identify biochemical processes affected by aging. Proteins of interest were identified as those contributing most to the variance in principal component analysis and those showing the largest significant differences between ages. These proteins implicated altered ECM composition, immune system function, and lipid metabolism.


Subject(s)
Bruch Membrane , Macular Degeneration , Animals , Mice , Bruch Membrane/chemistry , Proteome/analysis , Choroid , Macular Degeneration/metabolism , Aging/genetics
2.
Hum Mol Genet ; 32(2): 204-217, 2023 01 06.
Article in English | MEDLINE | ID: mdl-35943778

ABSTRACT

EFEMP1 R345W is a dominant mutation causing Doyne honeycomb retinal dystrophy/malattia leventinese (DHRD/ML), a rare blinding disease with clinical pathology similar to age-related macular degeneration (AMD). Aged Efemp1  R345W/R345W knock-in mice (Efemp1ki/ki) develop microscopic deposits on the basal side of retinal pigment epithelial cells (RPE), an early feature in DHRD/ML and AMD. Here, we assessed the role of alternative complement pathway component factor B (FB) in the formation of these deposits. RNA-seq analysis of the posterior eyecups revealed increased unfolded protein response, decreased mitochondrial function in the neural retina (by 3 months of age) and increased inflammatory pathways in both neural retina and posterior eyecups (at 17 months of age) of Efemp1ki/ki mice compared with wild-type littermate controls. Proteomics analysis of eye lysates confirmed similar dysregulated pathways as detected by RNA-seq. Complement activation was increased in aged Efemp1ki/ki eyes with an approximately 2-fold elevation of complement breakdown products iC3b and Ba (P < 0.05). Deletion of the Cfb gene in female Efemp1ki/ki mice partially normalized the above dysregulated biological pathway changes and oral dosing of a small molecule FB inhibitor from 10 to 12 months of age reduced sub-RPE deposits by 65% (P = 0.029). In contrast, male Efemp1ki/ki mice had fewer sub-RPE deposits than age-matched females, no elevation of ocular complement activation and no effect of FB inhibition on sub-RPE deposits. The effects of FB deletion or inhibition on Efemp1ki/ki mice supports systemic inhibition of the alternative complement pathway as a potential treatment of dry AMD and DHRD/ML.


Subject(s)
Macular Degeneration , Optic Disk Drusen , Male , Mice , Female , Animals , Complement Factor B/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Optic Disk Drusen/pathology , Retina/pathology , Retinal Pigment Epithelium/pathology
3.
Sci Rep ; 11(1): 10416, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001980

ABSTRACT

The complement system plays a role in the formation of sub-retinal pigment epithelial (RPE) deposits in early stages of age-related macular degeneration (AMD). But the specific mechanisms that connect complement activation and deposit formation in AMD patients are unknown, which limits the development of efficient therapies to reduce or stop disease progression. We have previously demonstrated that C3 blockage prevents the formation of sub-RPE deposits in a mouse model of EFEMP1-associated macular degeneration. In this study, we have used double mutant Efemp1R345W/R345W:C5-/- mice to investigate the role of C5 in the formation of sub-RPE deposits in vivo and in vitro. The data revealed that the genetic ablation of C5 does not eliminate the formation of sub-RPE deposits. Contrarily, the absence of C5 in RPE cultures promotes complement dysregulation that results in increased activation of C3, which likely contributes to deposit formation even in the absence of EFEMP1-R345W mutant protein. The results also suggest that genetic ablation of C5 alters the extracellular matrix turnover through an effect on matrix metalloproteinases in RPE cell cultures. These results confirm that C3 rather than C5 could be an effective therapeutic target to treat early AMD.


Subject(s)
Complement C5/metabolism , Extracellular Matrix Proteins/genetics , Retinal Degeneration/immunology , Retinal Pigment Epithelium/pathology , Animals , Complement Activation/genetics , Complement C5/genetics , Disease Models, Animal , Extracellular Matrix/metabolism , Female , Humans , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , Mutation , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Pigment Epithelium/immunology
4.
Prog Retin Eye Res ; 76: 100802, 2020 05.
Article in English | MEDLINE | ID: mdl-31704338

ABSTRACT

The ocular lens is a unique tissue that contains an age gradient of cells and proteins ranging from newly differentiated cells containing newly synthesized proteins to cells and proteins that are as old as the organism. Thus, the ocular lens is an excellent model for studying long-lived proteins (LLPs) and the effects of aging and post-translational modifications on protein structure and function. Given the architecture of the lens, with young fiber cells in the outer cortex and the oldest cells in the lens nucleus, spatially-resolved studies provide information on age-specific protein changes. In this review, experimental strategies and proteomic methods that have been used to examine age-related and cataract-specific changes to the human lens proteome are described. Measured spatio-temporal changes in the human lens proteome are summarized and reveal a highly consistent, time-dependent set of modifications observed in transparent human lenses. Such measurements have led to the discovery of cataract-specific modifications and the realization that many animal systems are unsuitable to study many of these modifications. Mechanisms of protein modifications such as deamidation, racemization, truncation, and protein-protein crosslinking are presented and the implications of such mechanisms for other long-lived proteins in other tissues are discussed in the context of age-related neurological diseases. A comprehensive understanding of LLP modifications will enhance our ability to develop new therapies for the delay, prevention or reversal of age-related diseases.


Subject(s)
Aging/metabolism , Cataract/metabolism , Crystallins/analysis , Lens, Crystalline/metabolism , Proteome/metabolism , Proteomics/methods , Animals , Humans
5.
Aging Cell ; 18(6): e13011, 2019 12.
Article in English | MEDLINE | ID: mdl-31385385

ABSTRACT

Late-onset retinal degeneration (L-ORD) is an autosomal dominant macular degeneration characterized by the formation of sub-retinal pigment epithelium (RPE) deposits and neuroretinal atrophy. L-ORD results from mutations in the C1q-tumor necrosis factor-5 protein (CTRP5), encoded by the CTRP5/C1QTNF5 gene. To understand the mechanism underlying L-ORD pathology, we used a human cDNA library yeast two-hybrid screen to identify interacting partners of CTRP5. Additionally, we analyzed the Bruch's membrane/choroid (BM-Ch) from wild-type (Wt), heterozygous S163R Ctrp5 mutation knock-in (Ctrp5S163R/wt ), and homozygous knock-in (Ctrp5S163R/S163R ) mice using mass spectrometry. Both approaches showed an association between CTRP5 and HTRA1 via its C-terminal PDZ-binding motif, stimulation of the HTRA1 protease activity by CTRP5, and CTRP5 serving as an HTRA1 substrate. The S163R-CTRP5 protein also binds to HTRA1 but is resistant to HTRA1-mediated cleavage. Immunohistochemistry and proteomic analysis showed significant accumulation of CTRP5 and HTRA1 in BM-Ch of Ctrp5S163R/S163R and Ctrp5S163R/wt mice compared with Wt. Additional extracellular matrix (ECM) components that are HTRA1 substrates also accumulated in these mice. These results implicate HTRA1 and its interaction with CTRP5 in L-ORD pathology.


Subject(s)
Collagen/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , Mutation , Retinal Degeneration/genetics , Animals , Cellular Senescence/genetics , Collagen/metabolism , High-Temperature Requirement A Serine Peptidase 1/metabolism , Humans , Mass Spectrometry , Mice , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
6.
Nat Protoc ; 11(7): 1206-18, 2016 07.
Article in English | MEDLINE | ID: mdl-27281648

ABSTRACT

Mouse models are powerful tools for the study of ocular diseases. Alterations in the morphology and function of the retinal pigment epithelium (RPE) are common features shared by many ocular disorders. We report a detailed protocol to collect, seed, culture and characterize RPE cells from mice. We describe a reproducible method that we previously developed to collect and culture murine RPE cells on Transwells as functional polarized monolayers. The collection of RPE cells takes ∼3 h, and the cultures mimic in vivo RPE cell features within 1 week. This protocol also describes methods to characterize the cells on Transwells within 1-2 weeks by transmission and scanning electron microscopy (TEM and SEM, respectively), immunostaining of vibratome sections and flat mounts, and measurement of transepithelial electrical resistance. The RPE cell cultures are suitable to study the biology of the RPE from wild-type and genetically modified strains of mice between the ages of 10 d and 12 months. The RPE cells can also be manipulated to investigate molecular mechanisms underlying the RPE pathology in the numerous mouse models of ocular disorders. Furthermore, modeling the RPE pathology in vitro represents a new approach to testing drugs that will help accelerate the development of therapies for vision-threatening disorders such as macular degeneration (MD).


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Retinal Pigment Epithelium/cytology , Animals , Cells, Cultured , Electric Impedance , Immunohistochemistry/methods , Mice , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Transmission/methods , Retinal Pigment Epithelium/ultrastructure
7.
Exp Eye Res ; 149: 59-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27339748

ABSTRACT

In the human ocular lens it is now realized that post-translational modifications can alter protein function and/or localization in fiber cells that no longer synthesize proteins. The specific sites of post-translational modification to the abundant ocular lens membrane proteins AQP0 and MP20 have been previously identified and their functional effects are emerging. To further understand how changes in protein function and/or localization induced by these modifications alter lens homeostasis, it is necessary to determine the spatial distributions of these modifications across the lens. In this study, a quantitative LC-MS approach was used to determine the spatial distributions of phosphorylated AQP0 and MP20 peptides from manually dissected, concentric layers of fiber cells from young and aged human lenses. The absolute amounts of phosphorylation were determined for AQP0 Ser235 and Ser229 and for MP20 Ser170 in fiber cells from the lens periphery to the lens center. Phosphorylation of AQP0 Ser229 represented a minor portion of the total phosphorylated AQP0. Changes in spatial distributions of phosphorylated APQ0 Ser235 and MP20 Ser170 correlated with regions of physiological interest in aged lenses, specifically, where barriers to water transport and extracellular diffusion form.


Subject(s)
Aging/metabolism , Aquaporins/metabolism , Eye Proteins/metabolism , Membrane Proteins/metabolism , Peroxiredoxins/metabolism , Adolescent , Adult , Blotting, Western , Chromatography, Liquid , Humans , Lens, Crystalline/metabolism , Middle Aged , Phosphorylation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Young Adult
8.
Adv Exp Med Biol ; 854: 53-8, 2016.
Article in English | MEDLINE | ID: mdl-26427393

ABSTRACT

Age related macular degeneration (AMD) is the primary cause of vision loss in the western world (Friedman et al., Arch Ophthalmol 122:564-572, 2004). The first clinical indication of AMD is the presence of drusen. However, with age and prior to the formation of drusen, extracellular basal deposits accumulate between the retinal pigment epithelium (RPE) and Bruch's membrane (BrM). Many studies on the molecular composition of the basal deposits and drusen have demonstrated the presence of extracellular matrix (ECM) proteins, complement components and cellular debris. The evidence reviewed here suggests that alteration in RPE cell function might be the primary cause for the accumulation of ECM and cellular debri found in basal deposits. Further studies are obviously needed in order to unravel the specific pathways that lead to abnormal formation of ECM and complement activation.


Subject(s)
Bruch Membrane/metabolism , Extracellular Matrix/metabolism , Macular Degeneration/metabolism , Retinal Drusen/metabolism , Retinal Pigment Epithelium/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Signal Transduction
9.
Hum Mol Genet ; 24(19): 5555-69, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26199322

ABSTRACT

Inherited and age-related macular degenerations (AMDs) are important causes of vision loss. An early hallmark of these disorders is the formation of sub-retinal pigment epithelium (RPE) basal deposits. A role for the complement system in MDs was suggested by genetic association studies, but direct functional connections between alterations in the complement system and the pathogenesis of MD remain to be defined. We used primary RPE cells from a mouse model of inherited MD due to a p.R345W mutation in EGF-containing fibulin-like extracellular matrix protein 1 (EFEMP1) to investigate the role of the RPE in early MD pathogenesis. Efemp1(R345W) RPE cells recapitulate the basal deposit formation observed in vivo by producing sub-RPE deposits in vitro. The deposits share features with basal deposits, and their formation was mediated by EFEMP1(R345W) or complement component 3a (C3a), but not by complement component 5a (C5a). Increased activation of complement appears to occur in response to an abnormal extracellular matrix (ECM), generated by the mutant EFEMP1(R345W) protein and reduced ECM turnover due to inhibition of matrix metalloproteinase 2 by EFEMP1(R345W) and C3a. Increased production of C3a also stimulated the release of cytokines such as interleukin (IL)-6 and IL-1B, which appear to have a role in deposit formation, albeit downstream of C3a. These studies provide the first direct indication that complement components produced locally by the RPE are involved in the formation of basal deposits. Furthermore, these results suggest that C3a generated by RPE is a potential therapeutic target for the treatment of EFEMP1-associated MD as well as AMD.


Subject(s)
Complement C3a/metabolism , Extracellular Matrix Proteins/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Retinal Pigment Epithelium/immunology , Animals , Cells, Cultured , Complement C5a/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Macular Degeneration/immunology , Matrix Metalloproteinases/metabolism , Mice , Mutation
10.
Hum Mol Genet ; 23(1): 52-68, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23943789

ABSTRACT

Macular degenerations, inherited and age related, are important causes of vision loss. Human genetic studies have suggested perturbation of the complement system is important in the pathogenesis of age-related macular degeneration. The mechanisms underlying the involvement of the complement system are not understood, although complement and inflammation have been implicated in drusen formation. Drusen are an early clinical hallmark of inherited and age-related forms of macular degeneration. We studied one of the earliest stages of macular degeneration which precedes and leads to the formation of drusen, i.e. the formation of basal deposits. The studies were done using a mouse model of the inherited macular dystrophy Doyne Honeycomb Retinal Dystrophy/Malattia Leventinese (DHRD/ML) which is caused by a p.Arg345Trp mutation in EFEMP1. The hallmark of DHRD/ML is the formation of drusen at an early age, and gene targeted Efemp1(R345W/R345W) mice develop extensive basal deposits. Proteomic analyses of Bruch's membrane/choroid and Bruch's membrane in the Efemp1(R345W/R345W) mice indicate that the basal deposits comprise normal extracellular matrix (ECM) components present in abnormal amounts. The proteomic analyses also identified significant changes in proteins with immune-related function, including complement components, in the diseased tissue samples. Genetic ablation of the complement response via generation of Efemp1(R345W/R345W):C3(-/-) double-mutant mice inhibited the formation of basal deposits. The results demonstrate a critical role for the complement system in basal deposit formation, and suggest that complement-mediated recognition of abnormal ECM may participate in basal deposit formation in DHRD/ML and perhaps other macular degenerations.


Subject(s)
Arginine/metabolism , Complement System Proteins/metabolism , Corneal Dystrophies, Hereditary/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Retinal Drusen/metabolism , Tryptophan/metabolism , Animals , Bruch Membrane/metabolism , Cell Adhesion , Complement C3/metabolism , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/immunology , Disease Models, Animal , Gene Knock-In Techniques , Humans , Mice , Mice, Transgenic , Optic Disk Drusen/congenital , Point Mutation , Proteomics , Reproducibility of Results , Retinal Drusen/pathology
11.
Exp Eye Res ; 93(6): 912-20, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22036630

ABSTRACT

Aquaporin-0 (AQP0), the major integral membrane protein in lens fiber cells, becomes highly modified with increasing age. The functional consequences of these modifications are being revealed, and the next step is to determine how these modifications affect the ocular lens, which is directly related to their abundances and spatial distributions. The aim of this study was to utilize matrix-assisted laser desorption ionization (MALDI) direct tissue profiling methods, which produce spatially-resolved protein profiles, to map and quantify AQP0 post-translational modifications (PTMs). Direct tissue profiling was performed using frozen, equatorial human lens sections of various ages prepared by conditions optimized for MALDI mass spectrometry profiling of membrane proteins. Modified forms of AQP0 were identified and further investigated using liquid chromatography tandem mass spectrometry (LC-MS/MS). The distributions of unmodified, truncated, and oleoylated forms of AQP0 were examined with a maximum spatial resolution of 500 µm. Direct tissue profiling of intact human lens sections provided high quality, spatially-resolved, relative quantitative information of AQP0 and its modified forms indicating that 50% of AQP0 is truncated at a fiber cell age of 24 ± 1 year in all lenses examined. Furthermore, direct tissue profiling also revealed previously unidentified AQP0 modifications including N-terminal acetylation and carbamylation. N-terminal acetylation appears to provide a protective effect against N-terminal truncation.


Subject(s)
Aquaporins/metabolism , Eye Proteins/metabolism , Lens, Crystalline/metabolism , Protein Processing, Post-Translational , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Acetylation , Adult , Age Factors , Aged , Aging/metabolism , Carbamates/metabolism , Child , Chromatography, Liquid , Humans , Oleic Acids/metabolism , Tandem Mass Spectrometry , Young Adult
12.
Ann N Y Acad Sci ; 1166: 27-37, 2009 May.
Article in English | MEDLINE | ID: mdl-19538261

ABSTRACT

Scrub typhus, caused by infection with Orientia tsutsugamushi, is probably the most common severe rickettsial disease. Early diagnosis followed by treatment with antibiotics such as doxycycline or chloramphenicol usually quickly decreases fever in patients, and they often recover well from other symptoms of the disease. However, poorly responsive cases have been reported from northern Thailand and southern India. In order to identify protein factors that may be partially responsible for differential drug sensitivity of isolates of Orientia, we compared the protein profiles of doxycycline sensitive (Karp) versus (vs.) insensitive (AFSC4 and AFSC7) isolates. Tryptic peptides from both total water-soluble proteins and from protein spots separated by 2D-PAGE were analyzed using LC-MS/MS. The identity of each protein was established using the published genomic sequence of Boryong strain O. tsutsugamushi. The profiles of protein released into water from these isolates were quite different. There were 10 proteins detected only in AFSC4, 3 only in Karp, and 1 only in AFSC7. Additionally, there were 2 proteins not detected only in AFSC4, 4 not found only in Karp, and 3 not found only in AFSC-7. A comparison of 2D-PAGE protein profiles of drug sensitive strain versus (vs.) insensitive isolates has led to the identification of 14 differentially expressed or localized proteins, including elongation factor Ts and Tu, DNA-directed RNA polymerase alpha-subunit, ATP synthase beta-subunit, and several hypothetical proteins. These data confirm the tremendous proteomic diversity of isolates of Orientia and suggest that drug insensitivity in this species may arise from multiple mechanisms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/analysis , Drug Resistance, Bacterial/physiology , Orientia tsutsugamushi , Proteome/analysis , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Bacterial Typing Techniques , Chromatography, Liquid , Doxycycline/therapeutic use , Electrophoresis, Gel, Two-Dimensional , Humans , Mass Spectrometry , Orientia tsutsugamushi/drug effects , Orientia tsutsugamushi/metabolism , Proteomics/methods , Scrub Typhus/drug therapy , Scrub Typhus/etiology , Scrub Typhus/microbiology
14.
Exp Eye Res ; 85(6): 861-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17935715

ABSTRACT

The present study was performed in order to obtain structural and quantitative information regarding the modifications that take place in the human lens as a result of tryptophan oxidation. In particular, the early tryptophan oxidation product, oxindolealanine (OIA) has been detected in lyophilized and hydrolyzed cataractous lenses by mass spectrometry. OIA was confirmed in human cataract samples by observing its ion (m/z 221), fragmentation pattern and absorption spectrum. Quantitative results indicate that there are differences in the amounts of OIA in the nucleus versus the cortex in human cataractous lenses. Expressed as a ratio to the level of phenylalanine (Phe), the nucleus has more than one and a half times greater levels of OIA as compared to the cortex [nucleus=(3.7+/-0.7)x10(-2) versus cortex=(2.3+/-0.3)x10(-2)]. Furthermore, the average value for the OIA/Phe ratio in the calf lens (controls) was (0.8+/-0.2)x10(-2) as compared to (3.7+/-0.7)x10(-2) in human cataractous lens nucleus (p<0.05). The quantitative results correspond to a 4.6-fold increase of OIA in human cataractous lenses. In a separate series of experiments using HPLC with photodiode array (PDA) detection only, the differences in OIA levels in cataract nucleus versus cortex and cataracts versus controls closely matched the LC/MS data. The results suggest that OIA levels are elevated in human cataractous lenses thus providing further evidence to implicate tryptophan oxidation in this process.


Subject(s)
Alanine/analogs & derivatives , Cataract/metabolism , Indoles/analysis , Lens, Crystalline/chemistry , Aged , Aged, 80 and over , Alanine/analysis , Animals , Biomarkers/analysis , Cattle , Chromatography, High Pressure Liquid/methods , Humans , Lens Cortex, Crystalline/chemistry , Lens Cortex, Crystalline/metabolism , Lens Nucleus, Crystalline/chemistry , Lens Nucleus, Crystalline/metabolism , Lens, Crystalline/metabolism , Oxidation-Reduction , Oxindoles , Spectrometry, Mass, Electrospray Ionization/methods , Spectrophotometry, Ultraviolet/methods , Tryptophan/metabolism , alpha-Crystallins/chemistry
15.
Hum Mol Genet ; 16(20): 2411-22, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17666404

ABSTRACT

Age-related macular degeneration (AMD) is the most common cause of vision loss in developed countries. A defining characteristic of this disorder is the accumulation of material between Bruch's membrane and the retinal pigment epithelium (RPE), first as microscopic basal deposits and later as clinically evident drusen. The pathogenesis of these deposits remains to be defined. Biochemical and genetic studies have suggested that inflammation and complement activation may play roles in AMD. Several lines of evidence also suggest that alterations to the extracellular matrix (ECM) of the RPE and choroid contribute to the development of AMD. The inherited macular degeneration Doyne honeycomb retinal dystrophy/Malattia Leventinese is thought to be caused by an R345W mutation in the EFEMP1 gene (also called fibulin-3). The pathogenicity of this mutation has been questioned because all individuals identified to date with the R345W mutation have shared a common haplotype. We investigated the pathogenicity of this mutation in families with early-onset macular degeneration and by generating Efemp1-R345W knockin mice. Genetic studies show that one of the identified families with the R345W mutation has a novel haplotype. The mutant Efemp1-R345W mice develop deposits of material between Bruch's membrane and the RPE, which resemble basal deposits in patients with AMD. These basal deposits contain Efemp1 and Timp3, an Efemp1 interacting protein. Evidence of complement activation was detected in the RPE and Bruch's membrane of the mutant mice. These results confirm that the R345W mutation in EFEMP1 is pathogenic. Further, they suggest that alterations in the ECM may stimulate complement activation, demonstrating a potential connection between these two etiologic factors in macular degeneration.


Subject(s)
Extracellular Matrix Proteins/genetics , Macular Degeneration/genetics , Point Mutation/physiology , Adolescent , Adult , Aged , Animals , Arginine/genetics , Child , DNA Mutational Analysis , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/physiology , Female , Humans , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Pedigree , Tryptophan/genetics
16.
FASEB J ; 21(12): 3318-28, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17548429

ABSTRACT

We have previously identified a gelsolin-like protein (C/L-gelsolin) as a corneal crystallin in zebrafish. Here we show by phylogenetic analysis that there are at least six genes encoding gelsolin-like proteins based on their gelsolin domains in zebrafish: gsna and gsnb group with the vertebrate gelsolin gene, scina and scinb group with the scinderin (adseverin) gene, and scinla (C/L-gelsolin) and scinlb are novel scinderin-like genes. RT-PCR showed that scinla, scinlb, and gsnb are preferentially expressed in the adult cornea whereas gsna is expressed to a similar extent in cornea, lens, brain, and heart; scina and scinb expression were detectable only in whole zebrafish and not in these adult tissues. Quantitative RT-PCR and 2-dimensional polyacrylamide gel electrophoresis followed by MALDI/TOF mass spectroscopy confirmed high expression of beta-actin and scinla, moderate expression of scinlb, and very low expression of gsna and gsnb in the cornea. Finally, transgenic zebrafish carrying a green fluorescent protein reporter transgene driven by a 4 kb scinla promoter fragment showed expression in the cornea, snout, dorsal fin, and tail fin of 3-day-old zebrafish larvae. Our data suggest that scinla and scinlb are diverged paralogs of the vertebrate scinderin gene and show that scinla encodes the zebrafish corneal crystallin previously called C/L-gelsolin.


Subject(s)
Cornea/chemistry , Crystallins/genetics , Gelsolin/genetics , Gene Duplication , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Animals, Genetically Modified , Crystallins/classification , Crystallins/metabolism , Gelsolin/classification , Gelsolin/metabolism , Humans , Microinjections , Molecular Sequence Data , Multigene Family , Phylogeny , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Distribution , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
17.
Cancer Immunol Immunother ; 56(5): 677-87, 2007 May.
Article in English | MEDLINE | ID: mdl-16967280

ABSTRACT

Within cancer research, phage display libraries have been widely used for the identification of tumor targeting peptides and antibodies. Additionally, phages are known to be highly immunogenic; therefore we evaluated the immunotherapeutic potential of tumor specific phages to treat established solid tumors in a mouse model of melanoma. We developed two tumor specific phages, one derived from a peptide phage display library and one Fab expressing phage with known specificity, for the treatment of mice bearing palpable B16-F10 or B16/A2K(b) tumors. Therapy in B16-F10 tumor bearing mice with tumor specific phages was superior to treatment with non-tumor specific phages and lead to delayed tumor growth and increased survival. In B16/A2K(b )tumor bearing mice, therapy with tumor specific phages resulted in complete tumor regression and long-term survival in 50% of the mice. Histological analysis of tumors undergoing treatment with tumor specific phages revealed that phage administration induced a massive infiltration of polymorphonuclear neutrophils. Furthermore, phages induced secretion of IL-12 (p70) and IFN-gamma as measured in mouse splenocyte culture supernatants. These results demonstrate a novel, immunotherapeutic cancer treatment showing that tumor specific phages can promote regression of established tumors by recruitment of inflammatory cells and induction of Th1 cytokines.


Subject(s)
Antigens, Neoplasm/immunology , Bacteriophages , Immunoglobulin Fab Fragments/immunology , Immunotherapy/methods , Melanoma, Experimental/therapy , Animals , Bacteriophages/genetics , Bacteriophages/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Library , Immunohistochemistry , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction
18.
Mol Cancer Res ; 5(12): 1225-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18171979

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with proinflammatory, proangiogenic, and protumorigenic properties. The molecular mechanisms underlying the role of MIF in tumorigenesis and angiogenesis are not well understood. To address these roles, an interfering MIF (iMIF) RNA was stably introduced into the B16-F10 mouse melanoma cell line, reducing MIF mRNA expression 1.6-fold and MIF protein expression 2.8-fold relative to control cells. When iMIF cells were subcutaneously injected into C57BL/6 mice, tumor establishment was significantly delayed and there was a marked absence of intratumoral vasculature in iMIF tumors relative to controls. A comparative gene expression analysis of iMIF and control melanoma cell lines revealed that thrombospondin-1 (TSP-1) mRNA expression was up-regulated 88-fold in the iMIF cells by real-time PCR. A 2-fold increase in TSP-1 protein levels was observed in iMIF cell culture supernatants. These results strongly suggest that the delayed tumor establishment and reduced vasculature in iMIF melanomas are linked to the up-regulation of the antiangiogenic TSP-1. They further define a novel function of MIF as a regulator of TSP-1 in a mouse melanoma model.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Melanoma/metabolism , Melanoma/pathology , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology , Thrombospondin 1/genetics , Animals , Cell Division , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Melanocytes/physiology , Melanoma/blood supply , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , Soft Tissue Neoplasms/blood supply , Subcutaneous Tissue , Thrombospondin 1/metabolism , Up-Regulation
19.
J Proteome Res ; 5(6): 1332-43, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16739985

ABSTRACT

The B16-F10 mouse model of melanoma is a widely used model to study many aspects of cancer biology and therapeutics in a solid tumor. Melanomas aggressively progress within a dynamic microenvironment containing in addition to tumor cells, stroma cells and components such as fibroblasts, immune cells, vascular cells, extracellular matrix (ECM) and extracellular molecules. The goal of this study was to elucidate the processes of tumor progression by identifying differentially expressed proteins in the tumor mass during specific stages of tumor growth. A comparative proteome analysis was performed on B16-F10 derived tumors in C57BL/6 mice at days 3, 5, 7, and 10. Statistical approaches were used to determine quantitative differential protein expression at each tumor time stage. Hierarchical clustering of 44 protein spots (p < 0.01) revealed a progressive change in the tumor mass when all 4 time stages were classified together, but there was a clear switch in expression of these proteins between the day 5 and the day 7 tumors. A trend analysis showed 53 protein spots (p < 0.001) following 6 predominant kinetic paths of expression as the tumor progressed. The protein spots were then identified using MALDI-TOF mass spectrometry. Proteins involved in glycolysis, inflammation, wounding, superoxide metabolism, and chemotaxis increased during tumorigenesis. From day 3 to day 7 VEGF and active cathepsin D were induced 7-fold and 4-fold, respectively. Proteins involved in electron transport, protein folding, blood coagulation, and transport decreased during tumorigenesis. This work illustrates changes in the biology of the B16-F10 tumor mass during tumor progression.


Subject(s)
Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Animals , Cathepsin D/metabolism , Electrophoresis, Gel, Two-Dimensional , Endothelial Cells/pathology , Female , Macrophages/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vascular Endothelial Growth Factor A/metabolism
20.
Biochemistry ; 43(30): 9856-65, 2004 Aug 03.
Article in English | MEDLINE | ID: mdl-15274640

ABSTRACT

Because of the lack of protein turnover in fiber cells of the ocular lens, Aquaporin 0 (AQP0), the most abundant membrane protein in the lens, undergoes extensive post-translational modification with fiber cell age. To map the distribution of modified forms of AQP0 within the lens, normal human lenses ranging in age from 34 to 38 were concentrically dissected into several cortical and nuclear sections. Membrane proteins still embedded in the membranes were digested with trypsin, and the resulting C-terminal peptides of AQP0 were analyzed by HPLC tandem mass spectrometry, permitting the identification of modifications and estimation of their abundance. Consistent with earlier reports, the major phosphorylation site was Ser 235, and the major sites of backbone cleavage occurred at residues 246 and 259. New findings suggest that cleavage at these sites may be a result of nonenzymatic truncation at asparagine residues. In addition, this approach revealed previously undetected sites of truncation at residues 249, 260, 261, and 262; phosphorylation at Ser 231 and to a lower extent at Ser 229; and racemization/isomerization of l-Asp 243 to d-Asp and d-iso-Asp. The spatial distribution of C-terminally modified AQP0 within the lens indicated an increase in truncation and racemization/isomerization with fiber cell age, whereas the level of Ser 235 phosphorylation increased from the outer to inner cortex but decreased in the nucleus. Furthermore, the remarkably similar pattern and distribution of truncation products from lenses from three donors suggest specific temporal mechanisms for the modification of AQP0.


Subject(s)
Cellular Senescence , Eye Proteins/chemistry , Eye Proteins/metabolism , Lens, Crystalline/chemistry , Lens, Crystalline/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Protein Processing, Post-Translational , Adult , Amino Acid Sequence , Aquaporins , Asparagine/metabolism , Cellular Senescence/physiology , Chromatography, Liquid , Eye Proteins/isolation & purification , Humans , Hydrolysis , Lens, Crystalline/cytology , Male , Membrane Glycoproteins/isolation & purification , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Phosphorylation , Protein Conformation , Sequence Deletion , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...