Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Exp Eye Res ; 204: 108471, 2021 03.
Article in English | MEDLINE | ID: mdl-33516764

ABSTRACT

PURPOSE: Complement activation is associated with choroidal neovascularization (CNV) in age-related macular degeneration (AMD). Fibroblast growth factor 2 (FGF2) and membrane attack complex (MAC) are present in eyes of patients with CNV. Herein, we investigated the effect of complement activation on FGF2 release in human retinal pigment epithelial (RPE) cells. METHODS: Cultured human RPE cells were primed with an anti-RPE antibody and then treated with C1q-depleted human serum in the presence or absence of Tec kinases inhibitor (LFM-A13). 38 cytokines/chemokines levels were measured by Luminex technology. Secretion of FGF2 and interleukin (IL)-6 was assessed by ELISA. Tec protein was measured by Western blot. mRNA expression of FGF2, chemokine (C-X-C motif) ligand 1 (CXCL-1), and family members of Tec kinases was evaluated by qPCR. Cell viability and MAC deposition were determined by WST-1 assay and flow cytometry, respectively. RESULTS: Complement activation caused increased FGF2 and IL-6 release. FGF2 was released when C6-depleted human serum was reconstituted with C6. Anti-C5 antibody significantly attenuated complement-mediated FGF2 release, but not IL-6. FGF2 mRNA levels were not affected, while CXCL-1 mRNA levels were increased by complement activation. FGF2-containing extracellular vesicles were detected in response to complement challenge. Tec mRNA and protein were expressed in RPE cells. In the presence of LFM-A13, secretion of FGF2, but not IL-6, and MAC deposition were significantly decreased and cell viability was significantly increased in complement-treated cells when compared to controls. CONCLUSIONS: Complement plays an important role to release FGF2 from RPE cells. Tec kinase is involved in MAC formation and complement-mediated FGF2 release. This information suggests a role for complement activation to mediate neovascularization in conditions such as AMD, and may elucidate potential therapeutic targets.


Subject(s)
Complement Activation/physiology , Complement System Proteins/metabolism , Fibroblast Growth Factor 2/metabolism , Retinal Pigment Epithelium/metabolism , Blotting, Western , Cells, Cultured , Choroidal Neovascularization/metabolism , Complement Membrane Attack Complex/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factor 2/genetics , Flow Cytometry , Humans , Interleukin-6/metabolism , Macular Degeneration/metabolism , Male , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
2.
Transl Vis Sci Technol ; 9(9): 37, 2020 08.
Article in English | MEDLINE | ID: mdl-32908800

ABSTRACT

Purpose: Factor I (FI) is a serine protease regulator of the complement system. Genetic variants in CFI are associated with advanced age-related macular degeneration (AAMD). However, the clinical and functional impact of these variants is unknown. This study assessed the functional significance of rare CFI variants using a serum-based assay. Methods: Carriers of rare variants with (n = 78) and without AAMD (n = 28), and noncarriers with (n = 49) and without AMD (n = 44) were evaluated. Function of FI was determined by measuring the proteolytic cleavage of C3b to iC3b, using the cofactor protein, Factor H. Results: CFI variants were categorized into three groups based on antigenic and functional assessments. Type 1 variants (n = 18) in 35 patients with AAMD demonstrated low serum FI levels and a corresponding decrease in FI function. Type 2 variants (n = 6) in 7 individuals demonstrated normal serum FI antigenic levels but reduced degradation of C3b to iC3b. Type 3 variants (n = 15) in 64 individuals demonstrated normal antigenic levels and degradation of C3b to iC3b. However, iC3b generation was low when measured per unit of FI. Thus most rare CFI variants demonstrate either low antigenic levels (type 1) or normal levels but reduced function (types 2 or 3). Conclusions: Results provide for the first time a comprehensive functional assessment in serum of CFI rare genetic variants and further establish FI's key role in the pathogenesis of AAMD. Translational Relevance: Stratifying patients in the clinic with a rare CFI variant will facilitate screening and targeting patients most likely to benefit from complement therapies.


Subject(s)
Complement Factor I , Macular Degeneration , Complement C3b , Complement Factor I/genetics , Complement System Proteins , Heterozygote , Humans , Macular Degeneration/genetics
3.
Invest Ophthalmol Vis Sci ; 58(7): 3073-3085, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28632844

ABSTRACT

PURPOSE: Complement activation is implicated in the pathogenesis of age-related macular degeneration (AMD). Apolipoprotein E (ApoE) and complement activation products such as membrane attack complex (MAC) are present in eyes of individuals with AMD. Herein, we investigated the effect of complement activation on induction of ApoE accumulation in human retinal pigment epithelial (RPE) cells. METHODS: Cultured human RPE cells were primed with a complement-fixing antibody followed by treatment with C1q-depleted (C1q-Dep) human serum to elicit alternative pathway complement activation. Controls included anti-C5 antibody-treated serum and heat-inactivated C1q-Dep. Total protein was determined on RPE cell extracts, conditioned media, and extracellular matrix (ECM) by Western blot. ApoE and MAC colocalization was assessed on cultured RPE cells and human eyes by immunofluorescent stain. ApoE mRNA expression was evaluated by quantitative PCR (qPCR). RESULTS: Complement challenge upregulated cell-associated ApoE, but not apolipoprotein A1. ApoE accumulation was blocked by anti-C5 antibody and enhanced by repetitive complement challenge. ApoE mRNA levels were not affected by complement challenge. ApoE was frequently colocalized with MAC in complement-treated cells and drusen from human eyes. ApoE was released into complement-treated conditioned media after a single complement challenge and accumulated on ECM after repetitive complement challenge. CONCLUSIONS: Complement challenge induces time-dependent ApoE accumulation in RPE cells. An understanding of the mechanisms by which complement affects RPE ApoE accumulation may help to better explain drusen composition, and provide insights into potential therapeutic targets.


Subject(s)
Apolipoproteins E/genetics , Complement Activation/physiology , Gene Expression Regulation , Macular Degeneration/genetics , RNA/genetics , Retinal Pigment Epithelium/metabolism , Apolipoproteins E/biosynthesis , Blotting, Western , Cells, Cultured , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mass Spectrometry , Middle Aged , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/pathology
4.
Invest Ophthalmol Vis Sci ; 55(5): 3012-21, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24677108

ABSTRACT

PURPOSE: Retinal pigment epithelial (RPE) cell death is an important feature of the advanced forms of AMD. Complement alternative pathway (AP) activation is associated with RPE cell death in AMD. In this study, we developed a new model to initiate AP activation on RPE cells and investigated the cellular mechanisms modulating AP activation-mediated RPE cell death. METHODS: An anti-RPE antibody was developed. A spontaneously arising human RPE cell line (ARPE-19) and donor RPE cells were primed with this antibody followed by stimulation with 6% C1q-depleted human serum (C1q-Dep) to activate AP. Complement activation was evaluated by flow cytometry and immunofluorescent staining. Cellular response to complement activation was examined by measurement of intracellular calcium and adenosine triphosphate (ATP) release. Cell viability was assessed by Sytox orange, tetrazolium salt, and lactate dehydrogenase release assays. RESULTS: Alternative pathway complement-mediated RPE cell death was associated with membrane attack complex formation and a rapid rise in intracellular calcium followed by release of ATP. Downregulation of membrane complement regulatory proteins and protein kinase C (PKC) inhibition increased cell susceptibility to complement attack. Pretreatment of RPE cells with either hydrogen peroxide or hydroquinone enhanced cell death. Chronic repetitive treatment of RPE cells with low levels of oxidants also enhanced complement-mediated cell death. CONCLUSIONS: Activation of complement through the alternative pathway induces sublytic and lytic phases of complement attack on RPE cells, leading to cell death modulated by extracellular calcium, membrane complement regulatory proteins, and intracellular signaling mechanisms. Single-dose oxidant exposure and low-dose repetitive oxidant exposure rendered RPE cells more susceptible to complement-mediated death.


Subject(s)
Calcium/metabolism , Cell Death/physiology , Complement Pathway, Alternative/physiology , Oxidative Stress/physiology , Protein Kinase C/physiology , Retinal Pigment Epithelium/physiology , Antibodies/pharmacology , Cell Line , Complement System Proteins/metabolism , Humans , Macular Degeneration , Membrane Proteins/metabolism , Retinal Pigment Epithelium/immunology
5.
Invest Ophthalmol Vis Sci ; 54(4): 2669-77, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23518773

ABSTRACT

PURPOSE: Retinal pigment epithelial (RPE) cell death occurs early in the pathogenesis of age-related macular degeneration (AMD) and Stargardt's disease. Emerging evidence suggests that all-trans-retinal (atRal) and alternative complement pathway (AP) activation contribute to RPE cell death in both of these retinal disorders. The aim of this study was to investigate the combined effect of atRal and AP activation on RPE cell viability. METHODS: RPE cells were treated with atRal and then incubated with a complement-fixing antibody followed by stimulation with C1q-depleted serum to activate AP. Cell viability was assessed by tetrazolium salt and lactate dehydrogenase release assays. Changes in cell surface CD46 and CD59 expression were assessed by flow cytometry. Cells were pretreated with the antioxidant resveratrol, and C1q-depleted serum was incubated with an anti-C5 antibody prior to initiating AP attack to determine the protective effects of antioxidant therapy and complement inhibition, respectively. RESULTS: Both atRal and AP activation independently caused RPE cell death. When AP attack was initiated following atRal treatment, a synergistic increase in cell death was observed. Following 24-hour atRal treatment, CD46 and CD59 expression decreased, corresponding temporally to increased susceptibility to AP attack. Resveratrol and the anti-C5 antibody both protected against AP-induced cell death following atRal exposure and were most effective when used in combination. CONCLUSIONS: atRal sensitizes RPE cells to AP attack, which may be mediated in part by atRal-induced downregulation of CD46 and CD59. Despite increased susceptibility to AP attack following exposure to atRal, resveratrol and anti-C5 antibody effectively prevent AP-mediated cell death.


Subject(s)
Complement Pathway, Alternative/immunology , Macular Degeneration/pathology , Retinal Pigment Epithelium/pathology , Angiogenesis Inhibitors/pharmacology , Cell Death/drug effects , Cell Death/immunology , Cells, Cultured , Flow Cytometry , Humans , Macular Degeneration/drug therapy , Macular Degeneration/immunology , Male , Middle Aged , Resveratrol , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/immunology , Ribonucleotide Reductases/antagonists & inhibitors , Stilbenes/pharmacology
6.
Proc Natl Acad Sci U S A ; 109(49): 20065-70, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23129651

ABSTRACT

An animal model of Leber hereditary optic neuropathy (LHON) was produced by introducing the human optic atrophy mtDNA ND6 P25L mutation into the mouse. Mice with this mutation exhibited reduction in retinal function by elecroretinogram (ERG), age-related decline in central smaller caliber optic nerve fibers with sparing of larger peripheral fibers, neuronal accumulation of abnormal mitochondria, axonal swelling, and demyelination. Mitochondrial analysis revealed partial complex I and respiration defects and increased reactive oxygen species (ROS) production, whereas synaptosome analysis revealed decreased complex I activity and increased ROS but no diminution of ATP production. Thus, LHON pathophysiology may result from oxidative stress.


Subject(s)
DNA, Mitochondrial/genetics , Disease Models, Animal , NADH Dehydrogenase/genetics , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/physiopathology , Oxidative Stress/physiology , Retina/pathology , Adenosine Triphosphate/metabolism , Age Factors , Animals , Demyelinating Diseases/etiology , Demyelinating Diseases/pathology , Electroretinography , Humans , Immunoblotting , Mice , Mutation, Missense/genetics , Optic Atrophy, Hereditary, Leber/complications , Optic Nerve/pathology , Reactive Oxygen Species/metabolism , Synaptosomes/metabolism
8.
J Biochem Biophys Methods ; 65(2-3): 107-20, 2005 Dec 31.
Article in English | MEDLINE | ID: mdl-16325916

ABSTRACT

Development of alphavbeta3-integrin inhibitors has been hampered by a lack of pharmacodynamic endpoints to identify doses that inhibit alphavbeta3 in vivo. To address this need, we developed an alphavbeta3 radioreceptor assay (RRA) that could be performed in 100% plasma. The RRA was based on 125I-echistatin binding to plate-immobilized alphavbeta3. Small molecule alphavbeta3 inhibitors efficiently competed echistatin binding to alphavbeta3 when the assay was carried out in buffer. However, when carried out in 100% plasma, the RRA revealed a 45 to >3000-fold loss in compound potencies. The losses in potency reflected, in part, the high plasma protein binding by the compounds examined. The RRA was adapted as an ex vivo pharmacodynamic model. Echistatin binding was measured in the presence of plasma harvested at timed intervals from rats dosed with select compounds. Using this pharmacodynamic model, compound and dose selection was optimized for further testing in models of corneal angiogenesis. Moderate anti-angiogenic activity was achieved when rats were dosed sufficient to achieve sustained (>50%) plasma inhibition through the trough interval. Thus, the RRA provided a simple technique to rank order compound potency in plasma, and could find general use as an ex vivo pharmacodynamic assay to select compounds and doses for preclinical and clinical proof-of-principle studies.


Subject(s)
Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/blood , Radioligand Assay/methods , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Proteins/metabolism , Cornea/blood supply , Cornea/drug effects , Drug Evaluation, Preclinical , In Vitro Techniques , Intercellular Signaling Peptides and Proteins , Male , Neovascularization, Pathologic/prevention & control , Peptides/blood , Peptides/pharmacokinetics , Protein Binding , Rats , Rats, Sprague-Dawley
9.
J Cell Sci ; 118(Pt 21): 4975-84, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16219679

ABSTRACT

Membrane type-1 matrix metalloproteinase (MT1-MMP) degrades the extracellular matrix, initiates the activation pathway of soluble MMPs and regulates the functionality of cell adhesion signaling receptors, thus playing an important role in many cell functions. Intracellular transport mechanisms, currently incompletely understood, regulate the presentation of MT1-MMP at the cell surface. We have focused our efforts on identifying these mechanisms. To understand the transport of MT1-MMP across the cell, we used substitution and deletion mutants, the trafficking of which was examined using antibody uptake and Chariot delivery experiments. Our experiments have demonstrated that the microtubulin cytoskeleton and the centrosomes (the microtubulin cytoskeleton-organizing centers) are essential for the trafficking and the internalization of MT1-MMP. We determined that after reaching the plasma membrane, MT1-MMP is internalized in the Rab-4-positive recycling endosomes and the Rab-11-positive pericentrosomal recycling endosomes. The microtubular trafficking causes the protease to accumulate in the pericentrosomal region of the cell. We believe that the presence of the transmembrane domain is required for the microtubular vesicular trafficking of MT1-MMP because the soluble mutants are not presented at the cell surface and they are not delivered to the centrosomes. The observed transport mechanisms provide a vehicle for the intracellular targets and, accordingly, for an intracellular cleavage function of MT1-MMP in malignant cells, which routinely overexpress this protease.


Subject(s)
Cell Membrane/enzymology , Metalloendopeptidases/metabolism , Microtubules/enzymology , Animals , Binding Sites, Antibody , CHO Cells , Cell Line , Cell Line, Tumor , Centrosome/enzymology , Centrosome/metabolism , Cricetinae , Cytoskeleton/physiology , Dogs , Humans , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metalloendopeptidases/immunology , Microtubules/metabolism , Protein Structure, Tertiary/physiology , Protein Transport/genetics , Protein Transport/physiology , Sequence Deletion , Transport Vesicles/enzymology , Transport Vesicles/genetics , Tubulin/physiology
10.
Traffic ; 5(8): 627-41, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15260832

ABSTRACT

Understanding the function of invasion-promoting membrane type-1 matrix metalloproteinase (MT1-MMP) is of paramount importance for understanding cancer biology. MT1-MMP is synthesized in cells as a latent zymogen that requires the cleavage of its prodomain to exert the proteolytic activity. The mature alphav integrin subunit is also generated by endoproteolytic cleavage of the alphav subunit precursor (pro-alphav). Cleavage by furin is considered to be a principal event in the activation of both MT1-MMP and pro-alphav. To elucidate the alternative activation pathway of MT1-MMP and pro-alphav, we employed furin-negative LoVo cells, which co-express MT1-MMP with integrin alphavbeta3. In these cells the MT1-MMP proenzyme was rapidly trafficked to the plasma membrane via an unconventional Brefeldin A-resistant pathway and, then, autocatalytically processed on the cell surface. Next, the MT1-MMP activity converted the cell surface-associated pro-alphav into the mature alphav integrin, represented by the disulfide-bonded heavy and light chains, and promoted the formation of the functional integrin alphavbeta3 heterodimer. These events stimulated cell motility in vitro, and malignant invasion and tumor growth in vivo. Our data suggest that in furin-negative colon carcinoma cells MT1-MMP is autocatalytically processed and the active protease then operates as a prointegrin convertase. Our findings argue strongly that the processing by furin is not a prerequisite for the activation of MT1-MMP.


Subject(s)
Colonic Neoplasms/metabolism , Furin/metabolism , Integrin alphaVbeta3/metabolism , Metalloendopeptidases/metabolism , Protein Precursors/metabolism , Protein Subunits/metabolism , Animals , Brefeldin A/metabolism , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Colonic Neoplasms/pathology , Cricetinae , Enzyme Activation , Furin/genetics , Humans , Matrix Metalloproteinase 14 , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/genetics , Mice , Mice, Mutant Strains , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Precursors/genetics , Protein Subunits/genetics , Protein Synthesis Inhibitors/metabolism , Protein Transport/physiology
11.
Exp Cell Res ; 291(1): 167-75, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14597417

ABSTRACT

We have recently demonstrated that in breast carcinoma MCF7 cells MT1-MMP processes the alphav, alpha3, and alpha5 integrin precursors generating the respective mature S-S-linked heavy and light alpha-chains. The precursor of alpha2 integrin subunit was found resistant to MT1-MMP proteolysis. The processing of the alphav subunit by MT1-MMP facilitated alphavbeta3-dependent adhesion, activation of FAK signaling pathway, and migration of MCF7 cells on vitronectin. To elucidate further the effects of MT1-MMP on cellular integrins, we examined the functional activity of alpha5beta1 and alpha2beta1 integrins in MCF7 cells expressing MT1-MMP. Either expression of MT1-MMP alone or its coexpression with alphavbeta3 failed to affect the functionality of alpha5beta1 integrin, and adhesion of cells to fibronectin. MT1-MMP, however, profoundly affected the cross-talk involving alphavbeta3 and alpha2beta1 integrins. In MT1-MMP-deficient cells, integrin alphavbeta3 suppressed the functional activity of the collagen-binding alpha2beta1 integrin receptor and diminished cell adhesion to type I collagen. Coexpression of MT1-MMP with integrin alphavbeta3 restored the functionality of alpha2beta1 integrin and, consequently, the ability of MCF7 cells to adhere efficiently to collagen. We conclude that the MT1-MMP-controlled cross-talk between alphavbeta3 and alpha2beta1 integrins supports binding of aggressive, MT1-MMP-, and alphavbeta3 integrin-expressing malignant cells on type I collagen, the most common substratum of the extracellular matrix.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Membrane/enzymology , Integrins/metabolism , Metalloendopeptidases/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Collagen Type I/metabolism , Enzyme Precursors/metabolism , Extracellular Matrix/metabolism , Female , Humans , Integrin alpha2beta1/metabolism , Integrin alphaVbeta3/metabolism , Matrix Metalloproteinases, Membrane-Associated , Neoplasm Metastasis , Protein Processing, Post-Translational , Protein Subunits/metabolism , Receptor Cross-Talk/physiology
12.
Invest Ophthalmol Vis Sci ; 43(4): 955-62, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11923234

ABSTRACT

PURPOSE: To determine in a corneal alkaline burn model of angiogenesis whether the expression of integrins and MMPs is consistent with a VEGF-induced angiogenic response. METHODS: Neovascularization in female Sprague-Dawley rats was induced by alkaline cauterization of the central cornea. RT-PCR for integrins alpha(1), alpha(2), beta(3), and beta(5); the endothelial marker CD31; and metalloproteinases MMP-2 and MT1-MMP was performed on naive corneas and on cauterized corneas 72 and 288 hours after cautery. Analyses of protein and MMP expression were conducted on naive corneas and on cauterized corneas 24, 72, 120, and 168 hours after cautery by immunofluorescence microscopy and gelatin zymography. RESULTS: RT-PCR indicated a correlation between the induced angiogenic response and the expression of alpha(1) and beta(3) integrin subunits and MT1-MMP. Immunohistochemical analysis indicated that alpha(1), alpha(2), alpha(5), and beta(5) integrins and MMP-2 and MT1-MMP were expressed on the newly developing vasculature. The beta(3) integrin was preferentially expressed on platelets. CONCLUSIONS: Integrin expression during neovascularization of rat corneas in response to alkaline injury correlates with an angiogenic response that uses the VEGF/alpha(v)beta(5) pathway. MMP-2 and MT1-MMP, but not MMP-9, are expressed in a pattern consistent with their involvement in the angiogenic response.


Subject(s)
Burns, Chemical/metabolism , Cornea/blood supply , Corneal Neovascularization/metabolism , Eye Burns/chemically induced , Integrins/metabolism , Matrix Metalloproteinase 2/metabolism , Metalloendopeptidases/metabolism , Alkalies , Animals , Burns, Chemical/etiology , Burns, Chemical/pathology , Cornea/pathology , Corneal Injuries , Corneal Neovascularization/etiology , Corneal Neovascularization/pathology , Female , Fluorescent Antibody Technique, Indirect , Integrins/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
13.
J Invest Dermatol ; 118(1): 147-54, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11851888

ABSTRACT

Squamous cell carcinoma antigen belongs to the serpin family and is used for the diagnosis and management of squamous cell carcinoma. We investigated the involvement of squamous cell carcinoma antigen in psoriasis, as it is always detected in the sera of patients with psoriasis. Squamous cell carcinoma antigen localization in psoriatic epidermis varied depending on its concentration in the patient's sera. When its level was low in serum, weak and scattered staining was observed in the granular layer. With a high concentration of squamous cell carcinoma antigen, strong staining through the suprabasal to granular layer and condensed staining around the plasma membrane or intracellular space was detected in the affected epidermis. Interestingly, squamous cell carcinoma antigen was abundant in nuclei of the granular layer cells and elongated rete ridges. Immunoelectron microscopy confirmed the localization of squamous cell carcinoma antigen in the nuclei as well as in the periphery of the cell membrane. A cDNA library was constructed from psoriatic epidermis and both clones, SCCA1 and SCCA2, were obtained. Attempts to raise specific antibodies or to prepare cRNA probes for SCCA1 and SCCA2 were unsuccessful because of their nearly identical structures. A primer pair from each reactive site sequence enabled us to give a distinctive product for SCCA1 and SCCA2 by reverse transcription polymerase chain reaction. Analysis using these primers demonstrated that the SCCA2 transcript was specifically expressed in psoriatic skin tissues. Our results suggest that overexpression of squamous cell carcinoma antigens is associated with the disease activity of psoriasis.


Subject(s)
Antigens, Neoplasm/metabolism , Psoriasis/metabolism , Serpins/metabolism , Skin/metabolism , Adult , Aged , Antigens, Neoplasm/genetics , Cell Nucleus/metabolism , Humans , In Situ Hybridization , Microscopy, Immunoelectron , Middle Aged , RNA, Messenger/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
14.
J Biol Chem ; 277(14): 12270-4, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11805099

ABSTRACT

Syndecan-4 and integrins are the primary transmembrane receptors of focal adhesions in cells adherent to extracellular matrix molecules. Syndesmos is a cytoplasmic protein that interacts specifically with the cytoplasmic domain of syndecan-4, and it co-localizes with syndecan-4 in focal contacts. In the present study we sought possible interactors with syndesmos. We find that syndesmos interacts with the focal adhesion adaptor protein paxillin. The binding of syndesmos to paxillin is direct, and these interactions are triggered by the activation of protein kinase C. Syndesmos also binds the paxillin homolog, Hic-5. The connection of syndecan-4 with paxillin through syndesmos parallels the connection between paxillin and integrins and may thus reflect the cooperative signaling of these two receptors in the assembly of focal adhesions and actin stress fibers.


Subject(s)
Carrier Proteins/chemistry , Cytoskeletal Proteins/chemistry , DNA-Binding Proteins/chemistry , Phosphoproteins/chemistry , Actins/metabolism , Amino Acid Sequence , Animals , Cell Adhesion , Cells, Cultured , Chick Embryo , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Glutathione Transferase/metabolism , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Models, Biological , Molecular Sequence Data , Paxillin , Phosphoproteins/metabolism , Precipitin Tests , Protein Binding , Protein Biosynthesis , Protein Kinase C/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Transcription, Genetic
15.
J Biol Chem ; 277(9): 7377-85, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11741954

ABSTRACT

Membrane type-1 matrix metalloproteinase (MT1-MMP) and alpha(v)beta(3) integrin are both essential to cell invasion. Maturation of integrin pro-alpha(v)chain (pro-alpha(v)) involves its cleavage by proprotein convertases (PC) to form the disulfide-bonded 125-kDa heavy and 25-kDa light alpha chains. Our report presents evidence of an alternative pathway of pro-alpha(v) processing involving MT1-MMP. In breast carcinoma MCF7 cells deficient in MT1-MMP, pro-alpha(v) is processed by a conventional furin-like PC, and the mature alpha(v) integrin subunit is represented by the 125-kDa heavy chain and the 25-kDa light chain commencing from the N-terminal Asp(891). In contrast, in cells co-expressing alpha(v)beta(3) and MT1-MMP, MT1-MMP functions as an integrin convertase. MT1-MMP specifically cleaves pro-alpha(v), generating a 115-kDa heavy chain with the truncated C terminus and a 25-kDa light chain commencing from the N-terminal Leu(892). PC-cleavable alpha(3) and alpha(5) but not the PC-resistant alpha(2) integrin subunit are also susceptible to MT1-MMP cleavage. These novel mechanisms involved in the processing of integrin alpha subunits underscore the significance and complexity of interactions between MT1-MMP and adhesion receptors and suggest that regulation of integrin functionality may be an important role of MT1-MMP in migrating tumor cells.


Subject(s)
Antigens, CD/metabolism , Metalloendopeptidases/chemistry , Amino Acid Sequence , Animals , Aspartic Acid/chemistry , Binding Sites , Blotting, Western , Cell Adhesion , Cell Movement , Cytoplasm/metabolism , Disulfides , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Integrin alphaV , Integrins/metabolism , Matrix Metalloproteinase 14 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptors, Vitronectin/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL