Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Arch Ophthalmol ; 118(9): 1269-76, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980774

ABSTRACT

OBJECTIVES: To report the clinical characteristics of a family with autosomal dominant retinitis pigmentosa caused by a proline-to-alanine mutation at codon 23 (Pro23Ala) of the rhodopsin gene and to compare this phenotype with that associated with the more common proline-to-histidine mutation at codon 23 (Pro23His). METHODS: We examined 6 patients within a single pedigree. The electroretinograms (ERGs) of 35 patients with known Pro23His mutations and of 22 healthy individuals were reviewed. Scotopic dim flash-response amplitudes, maximum combined-response amplitudes, and photopic-response amplitudes from the ERGs of these patients were plotted against age. The ERG indices of 5 individuals in the Pro23Ala family were compared with those of the patients with Pro23His mutations and of healthy individuals. Multiple linear regression was performed to evaluate the effect of age and mutation type on amplitudes. Mutation detection was performed using single-strand conformation polymorphism analysis, followed by automated DNA sequencing. RESULTS: Patients with the Pro23Ala mutation have a clinical phenotype characterized by onset of symptoms in the second to fourth decades of life, loss of superior visual field with relatively well-preserved inferior fields, and mild nyctalopia. Comparison with patients with the Pro23His mutation demonstrates statistically significant differences (P<.001) in responses to dim flash, maximum combined, and photopic responses between patients with these mutations after controlling for the effects of age. Patients with Pro23Ala mutations were less affected by ERG criteria than patients with Pro23His mutations. Patients with Pro23Ala mutations also differed significantly from healthy patients in all ERG indices examined (P<.001), after controlling for age. CONCLUSION: We describe a rare mutation in codon 23 of rhodopsin causing autosomal dominant retinitis pigmentosa. The retinal dystrophy associated with the Pro23Ala mutation is characteristically mild in presentation and course, with greater preservation of ERG amplitudes than the more prevalent Pro23His mutation. Arch Ophthalmol. 2000;118:1269-1276


Subject(s)
Electroretinography , Point Mutation , Retina/physiopathology , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Adolescent , Adult , Aged , Child , Codon , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Pedigree , Phenotype , Photic Stimulation , Polymorphism, Single-Stranded Conformational , Retina/pathology , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/physiopathology , Visual Acuity , Visual Field Tests , Visual Fields
2.
Invest Ophthalmol Vis Sci ; 41(1): 36-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634598

ABSTRACT

PURPOSE: To map a gene for cataracts in a family with congenital nuclear and sutural cataracts and to examine candidate genes in the linked region. METHODS: A large family with autosomal dominant congenital nuclear and sutural cataracts was identified and characterized. A genome-wide screen was conducted with a set of markers spaced at 10- to 15-cM intervals, and linkage was assessed using standard LOD score analysis. RESULT: Fifteen (15) affected individuals were identified. This form of congenital cataracts maps to a 12-cM region on chromosome 3q21.2-q22.3 between markers D3S3674 and D3S3612, with a maximum multipoint LOD score of 6.94 at D3S1273. The crystallin gene, CRYGS, was excluded as a candidate gene for this locus. CONCLUSIONS: There are now more than 12 different genetic loci that cause congenital cataracts. The most recent locus to be identified is on chromosome 3q21.2-q22.3, in a family with congenital nuclear and sutural cataracts.


Subject(s)
Cataract/congenital , Cataract/genetics , Chromosome Mapping , Chromosomes, Human, Pair 3/genetics , Adult , Child, Preschool , Crystallins/genetics , DNA/analysis , DNA Primers/chemistry , Female , Genetic Linkage , Humans , Male , Molecular Sequence Data , Pedigree
3.
J Lab Clin Med ; 133(5): 507-15, 1999 May.
Article in English | MEDLINE | ID: mdl-10235134

ABSTRACT

We report here the discovery of two novel human platelet and megakaryocytic DAMI cell enzymes that have beta-secretase-like activity. These activities could potentially effect cleavage of the amyloid precursor protein (APP) at the beta-amyloid peptide N-terminus, by an EC 3.4.24.15-like metalloprotease, and the N terminus-1 position, by a serine protease. Thus both enzymes may generate the amyloidogenic beta-peptide. Studies of intact and Triton X-100-lysed DAMI cells, as well as intact versus subcellular fractions of platelets, demonstrate the presence of these proteolytic activities. The resting platelet has (1) a surface serine protease, demonstrated by its ability to cleave a beta-secretase substrate and by its inhibitor sensitivity; and (2) a metalloprotease, recognized by an antibody to EC 3.4.24.15, which resides intracellularly in the alpha-granule membrane, is translocated to the surface on activation, and shows beta-secretase-like activity by cleaving the same substrate. This metalloprotease can also cleave recombinant APP to a potentially amyloidogenic fragment. Surface metalloprotease was identified in DAMI cells by flow cytometry and Western blotting with a specific anti-EC 3.4.24.15 monoclonal antibody, while activity was identified by using two beta-secretase substrates. This article is the first to document two previously unknown endoproteinases with beta-secretase-like activity in platelets and DAMI cells. These proteases are capable of effecting cleavage of APP and could therefore contribute to Abeta deposition in the cerebrovasculature.


Subject(s)
Alzheimer Disease/enzymology , Blood Platelets/enzymology , Endopeptidases/metabolism , Megakaryocytes/enzymology , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/metabolism , Antibodies, Monoclonal/immunology , Aspartic Acid Endopeptidases , Enzyme Inhibitors/pharmacology , Humans , Membrane Proteins/immunology , Metalloendopeptidases/metabolism , Naphthalenesulfonates/metabolism , Oligopeptides/metabolism , Peptides/metabolism , Serine Endopeptidases/metabolism , Substrate Specificity , Thrombin/metabolism , Tumor Cells, Cultured
4.
Amyloid ; 5(3): 153-62, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9818052

ABSTRACT

Proteolytic cleavage of the amyloid precursor protein (A beta PP) results in the generation of the amyloidogenic fragment known as amyloid beta peptide (A beta). Deposition of A beta in the brain parenchyma and cerebrovasculature is a feature of Alzheimer's disease (AD). To date, the process whereby A beta is generated and deposited remains unclear. We have previously established that activated platelets from AD patients retain more A beta PP on their surface than control platelets. We report here that an endothelial cell-derived enzyme can cleave this surface platelet A beta PP. Human blood brain barrier endothelial cells from brains of AD patients were assayed for potential A beta PP-cleaving enzymes using synthetic peptide substrates encompassing the A beta N-terminus cleavage site. A protease activity capable of cleaving A beta PP on the surface of AD platelets was noted. The A beta PP cleavage is partially inhibited by EDTA, by ZincOV, as well as by a specific inhibitor of the Zn metalloprotease E.C.3.4.24.15. Furthermore, the protease is recognized by an antibody directed against it, using immunohistochemistry, Western blot analysis and flow cytometry. The protease is not secreted, but rather resides intracellularly as well as on the surface of the endothelial cells. The data suggest that E.C.3.4.24.15 synthesized by brain endothelial cells may process the platelet-derived A beta PP, yielding fragments which could contribute to cerebrovascular A beta deposits.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/metabolism , Blood-Brain Barrier , Endopeptidases/isolation & purification , Endothelium, Vascular/enzymology , Metalloendopeptidases/isolation & purification , Amyloid Precursor Protein Secretases , Antibody Specificity , Aspartic Acid Endopeptidases , Blood Platelets/metabolism , Edetic Acid/pharmacology , Endopeptidases/immunology , Humans , Hydroxamic Acids/pharmacology , Immunohistochemistry , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/immunology , Oligopeptides/metabolism
5.
J Lab Clin Med ; 132(4): 341-50, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9794706

ABSTRACT

We have previously demonstrated that thrombin-activated platelets from patients with advanced Alzheimer's disease (AD) retain significantly more surface membrane-bound amyloid precursor protein (mAPP) than platelets from non-demented age-matched individuals (AM). We have studied interactions between these platelets and the cerebrovascular endothelium to which activated platelets adhere in a model system, investigating their involvement in the formation of amyloid beta peptide (Abeta) deposits in AD patients. We report here that there appear to be alpha and beta secretase-like activities in primary human blood brain barrier endothelial cell (BEC) cultures from both AD patients and AM control subjects (AD-BEC and AM-BEC, respectively) as well as a gamma secretase-like activity that appears only in AD-BEC. No such activities were observed in human umbilical vein endothelial cells (HUVECs). Furthermore, there is more penetration of the platelet-released products platelet factor 4 and soluble APP through the BEC layer grown from AD patients than that grown from AM individuals, whereas none penetrate through a HUVEC layer. Thus the interaction between platelets, the APP they have retained or released, and cerebral vascular endothelial cells may be at least partially responsible for amyloidogenic deposits around the cerebral vasculature of AD patients.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/metabolism , Brain , Endopeptidases/physiology , Endothelium, Vascular/enzymology , Adult , Aged , Aged, 80 and over , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Blood Platelets/metabolism , Blood-Brain Barrier , Blotting, Western , Brain/blood supply , Brain/enzymology , Endothelium, Vascular/cytology , Female , Humans , Male , Middle Aged , Umbilical Veins/cytology
6.
Infect Immun ; 66(1): 28-35, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9423835

ABSTRACT

CD14 is a signaling receptor for both gram-negative bacterial lipopolysaccharide (LPS) and mycobacterial lipoarabinomannan (LAM) that lacks terminal mannosyl units (AraLAM). In contrast, terminally mannosylated LAM (ManLAM) binds the macrophage mannose receptor (MMRc), although the ability of the MMRc to serve as a signaling receptor has not been previously reported. We compared the abilities of AraLAM and ManLAM to induce distinct responses in two monocytic cell populations, freshly isolated human peripheral blood monocytes (PBM) and monocyte-derived macrophages (MDM). The responses examined were chemotaxis and transient changes in free cytosolic calcium ([Ca2+]in). We found that AraLAM but not ManLAM was chemotactic for both PBM and MDM. Migration of these cells in vitro to AraLAM was specifically blocked by an anti-CD14 monoclonal antibody, suggesting that CD14 mediates the chemotactic response to AraLAM. Subsequently, we found that AraLAM induced a transient rise in [Ca2+]in levels within a subpopulation of PBM but not MDM. This response was blocked by anti-CD14 antibodies. In contrast, ManLAM induced a transient rise in [Ca2+]in levels within a subpopulation of MDM but not PBM. This response was blocked by either anti-CD14 or anti-MMRc antibodies. These data suggest that the MMRc can serve as a signaling receptor and that coligation of both CD14 and the MMRc is required to elicit a specific response. Thus, one response to LAM (chemotaxis) can be elicited solely by engaging CD14, whereas a different response (changes in [Ca2+]in levels) depends on both the differentiation state of the cells and concomitant engagement of CD14 and the MMRc.


Subject(s)
Lectins, C-Type , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/immunology , Macrophages/immunology , Mannose-Binding Lectins , Monocytes/immunology , Mycobacterium/immunology , Receptors, Cell Surface/physiology , Antibodies, Blocking , Antibodies, Monoclonal/immunology , Calcium/metabolism , Chemotaxis , Cytoplasm/metabolism , Down-Regulation , Flow Cytometry , Humans , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/isolation & purification , Macrophages/metabolism , Mannose Receptor , Monocytes/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Receptors, Cell Surface/immunology , Receptors, Complement/metabolism , Receptors, Complement/physiology , Signal Transduction/immunology
7.
J Lab Clin Med ; 130(1): 21-32, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9242363

ABSTRACT

Platelets, when released as anuclear cells by their precursor megakaryocytes, already carry soluble proteolytic fragments of the amyloid precursor protein (APP) within their alpha-granules and intact APP in the alpha-granule membranes. In response to activation signals elicited by physiologic stimuli such as thrombin, platelets release their granules' soluble contents and translocate granule membrane-bound proteins to the plasma membrane. Because platelets carry >90% of the circulation's APP, activated platelets have been implicated as origins of the beta-amyloid peptide fragment of APP (A beta), whose deposition in the cerebrovasculature is characteristic of Alzheimer's disease. We have therefore studied the APP contents and proteolytic processing in resting DAMI human megakaryocytic cells, along with the consequences of the activation of these cells by thrombin, comparing the results in each case to those with human platelets. Resting and PMA-differentiated DAMI cell contents were examined by Western blotting, immunoprecipitation, or metabolic labeling with sulfur 35-labeled methionine during culture, while plasma membrane-bound APP was evaluated by flow cytometry. Activation was followed by changes in cytoplasmic calcium concentration ((Ca++)in) and in membrane potential. Like platelets, DAMI cells exhibited a thrombin dose-dependent delta(Ca++)in, and membrane potential change; in contrast to the surface of a platelet, the surface of an agranular resting DAMI cell expresses granule-membrane proteins (APP and CD63) that appear on platelets only after activation. DAMI cell culture with 35S-labeled methionine confirmed that megakaryocytes synthesize large amounts of APP, of slightly higher molecular weight, and degrade their APP extensively before platelets are formed.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Blood Platelets/metabolism , Megakaryocytes/physiology , Blood Platelets/drug effects , Blotting, Western , Calcium/metabolism , Cell Line , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Flow Cytometry , Humans , Megakaryocytes/drug effects , Membrane Potentials , Signal Transduction/physiology , Thrombin/pharmacology
8.
Neurobiol Aging ; 18(2): 147-53, 1997.
Article in English | MEDLINE | ID: mdl-9258891

ABSTRACT

Upon activation, platelet alpha-granules' soluble contents are secreted and membrane-bound contents are translocated to the plasma membrane. Membrane-bound proteins include the beta-amyloid precursor protein (APP) from which the beta-amyloid (A beta) deposits found surrounding the cerebrovasculature of patients with Alzheimer's Disease (AD) may originate. We show here that activated platelets from AD patients exhibit less APP processing, retain more of the protein on their surface, and secrete less as soluble fragments than do controls. Surface labeling demonstrated that there is little APP or CD62 on the surface of resting platelets. Upon activation, control platelets exhibited more of both proteins on their surface, while advanced AD patients exhibited similar amounts of CD62 as controls, but retained significantly more surface APP. AD platelets secreted similar amounts of most soluble alpha-granule contents as controls, but less APP fragments. Together these results suggest a processing defect that may account for greater deposition of A beta-containing products in the vasculature to which activated platelets adhere.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Peptides/blood , Blood Platelets/metabolism , Adult , Aged , Amyloid beta-Protein Precursor/blood , Blotting, Western , Cell Degranulation , Cell Membrane/metabolism , Dementia/blood , Electrophoresis, Polyacrylamide Gel , Female , Humans , Male , Platelet Activation/physiology
9.
J Immunol Methods ; 209(2): 165-75, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9461332

ABSTRACT

Isolation of monocytes by plastic adherence alters cell morphology and function. In order to study the effects of cell isolation procedures and subsequent culture on monocyte function, we examined cytoplasmic calcium concentration changes (delta[Ca2+]in) in human monocytes isolated by either negative (magnetic bead) or positive (plastic adherence) selection then stimulated with formyl-Met-Leu-Phe (fMLP), either immediately after isolation, or after 48 h in culture. We have previously shown that fresh adherence-isolated monocytes respond to fMLP with small delta[Ca2+]in and oxidative burst responses, exhibiting larger responses following 48 h of incubation. We now demonstrate that fresh monocytes, prevented from adhering by negative selection, exhibit an even smaller fMLP-induced delta[Ca2+]in, which does not increase during 48 h in culture if cells are kept nonadherent, in Teflon. Calcium responses of adherent, fresh monocytes do not increase if cells are subsequently placed into suspension and maintained nonadherent, but increase if nonadherent cells are permitted to adhere to plastic. Furthermore, augmented fMLP-[Ca2+]in and oxidative burst responses in plastic-adherent cells are restricted to a CD14-high phenotype subpopulation. The CD14-high phenotype also describes a subpopulation of cells that responds to CD4 crosslinking with a rapid delta[Ca2+]in. Induction of a subpopulation of CD14-high expressing cells by adherence may explain in part maturation-induced response changes observed in macrophage but not in monocyte in vitro systems.


Subject(s)
Calcium/metabolism , Cell Adhesion , Lipopolysaccharide Receptors/immunology , Monocytes/immunology , Signal Transduction , CD4 Antigens/immunology , Flow Cytometry , Humans , Monocytes/drug effects , Monocytes/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Respiratory Burst
SELECTION OF CITATIONS
SEARCH DETAIL
...